COMBINATION CANCER THERAPIES

Information

  • Patent Application
  • 20230346743
  • Publication Number
    20230346743
  • Date Filed
    January 26, 2023
    a year ago
  • Date Published
    November 02, 2023
    6 months ago
Abstract
Drug combinations of a heteroarotinoid (e.g., SHetA2, SHetA3, SHetA4, SHetC2, SHetD3, SHetD4, SHetD5, SHet50, SHet65, SHet100, OHet72, NHet17, NHet86, or NHet90), and an Azabicyclooctan-3-one derivative (e.g., PRIMA-1 or PRIMAMET) and/or a CDK4/6 inhibitor (e.g., Palbociclib, Abemaciclib, Ribociclib, Narazaciclib (ON123300), Dalpiciclib, Dinaciclib, Milciclib, Seliciclib), which are effective as anti-cancer treatments, and kits and methods of use of such drug combinations.
Description
BACKGROUND

Despite being cancer free after primary cytoreductive surgery and platinum-based chemotherapy, approximately 70% of ovarian cancer patients will relapse after 3 years. Maintenance therapy using the FDA-approved PARP-1 inhibitor, olaparib, in BRCA mutation positive patients, or the angiogenesis inhibitor, bevacizumab, in all other patients, has been shown to prolong time to recurrence, however no improvement in overall survival was found in long term follow-up for bevacizumab or interim analysis of 21% data for olaparib. Both of these therapies have significant side effects that limit dosing and duration of treatment. Furthermore, olaparib caused an increased risk of developing secondary cancers.


A rational approach for developing non-toxic maintenance therapy for ovarian cancer is to restore normal p53 activity. Mutations in the TP53 gene occur with a 96 to 100% frequency in high-grade serous ovarian cancer (HGSOC), the most common and lethal type of ovarian cancer, which can arise from fallopian tube fimbriae. Mutation or loss of p53 protein can reduce cancer therapy response. Currently p53 restoration is a target of multiple anti-cancer drug strategies, including by way of reactivation of the wild type p53 functions in missense mutant p53 (m-p53) and release of p53 from inhibitory proteins. PRIMA-1 (“p53 re-activation and induction of massive apoptosis-1”) is a p53 reactivator that modifies amino acids in the m-p53 core domain to restore proper protein conformation and function. PRIMA-1MET (APR-246), a methylated analog of PRIMA-1, exhibited greater cancer cell line cytotoxicity and synergy with chemotherapy, and is undergoing clinical investigation. A first-in-human clinical trial found PRIMA-1MET to have a favorable pharmacokinetic profile, to induce wild type p53 molecular and cellular activities in tumors and to be safe at plasma levels predicted to have therapeutic effects. Currently, clinicaltrials.gov reports multiple ongoing trials evaluating PRIMA-1MET in combination with chemotherapy for several malignancies, including recurrent ovarian cancer.


Another drug in development that can induce p53 activity is SHetA2 (NSC 726189), an orally bioavailable, small molecule drug that disrupts mortalin/p53 complexes in ovarian cancer cells. This drug is currently being developed for clinical trials based on its induction of apoptosis in cancer cells, while its effect on healthy cells is limited to G1 cell cycle arrest. SHetA2 also exhibited chemoprevention activity. Extensive preclinical studies conducted by the US National Cancer Institute (NCI) RAID and RAPID Programs, and others, demonstrated that SHetA2 did not cause mutagenicity, teratogenicity or toxicity, and exhibited a pharmacologic profile suitable for an oral chemoprevention agent. Peer-reviewed NCI R01 and PREVENT grants are supporting first-in-human clinical trials of SHetA2 capsules.





BRIEF DESCRIPTION OF THE DRAWINGS

Several embodiments of the present disclosure are hereby illustrated in the appended drawings. It is to be noted however, that the appended drawings only illustrate several typical embodiments and are therefore not intended to be considered limiting of the scope of the inventive concepts disclosed herein. The patent or application file contains at least one drawing executed in color.



FIG. 1. Upregulation of mortalin expression in ovarian cancer. Representative images of mortalin immunohistochemical staining from the GOG Tissue Microarray (TMA) of ovarian cancer tumor progression.



FIG. 2. Upregulation of mortalin expression in ovarian cancer epithelial cells. Comparison of average mortalin immunohistochemical positivity stain scores for epithelial cells in benign, borderline and serous tumor tissues in the GOG Tissue Microarray (TMA) of ovarian cancer tumor progression. *p ≤ 0.05, ***p ≤ 0.001.



FIG. 3. Upregulation of mortalin expression in ovarian cancer. Comparison of average mortalin immunohistochemical positivity stain scores for stromal cells in benign, borderline and serous tumor tissues in the GOG Tissue Microarray (TMA) of ovarian cancer tumor progression. *p ≤ 0.05, ***p ≤ 0.001.



FIG. 4. SHetA2 causes p53 accumulation in the nucleus of cancer cells, but not healthy cells. Immunofluorescence assay was performed on A2780 and OV90 ovarian cancer cell lines and FTSECs (normal fallopian tube secretory epithelial cells) by detecting p53 with Alexa fluor 647 (Red) fluorochrome-conjugated antibody and nuclei were stained with DAPI (blue). Imaging was performed with confocal microscope at 63X.



FIG. 5A. SHetA2 causes p53 accumulation in the nucleus of cancer cells. Immunoblots of p53 in cytoplasmic and nuclear protein fractions A2780 ovarian cancer cells. GAPDH and Histone H3 served as loading controls for cytoplasmic and nuclear fractions, respectively.



FIG. 5B. SHetA2 causes p53 accumulation in the nucleus of cancer cells. Bar graphs of quantified p53 band densities, which were normalized to the loading controls of the western blots in FIG. 5A.



FIG. 6A. SHetA2 causes p53 accumulation in the nucleus of cancer cells. Immunoblots of p53 in cytoplasmic and nuclear protein fractions OV90 ovarian cancer cells. GAPDH and Histone H3 served as loading controls for cytoplasmic and nuclear fractions, respectively.



FIG. 6B. SHetA2 causes p53 accumulation in the nucleus of cancer cells. Bar graphs of quantified p53 band densities, which were normalized to the loading controls of the western blots in FIG. 6A.



FIG. 7A. Mitochondrial accumulation of p53 after SHetA2 treatment in cancer cells. p53 levels were examined in western blots of cytoplasmic and mitochondrial fractions of A2780 ovarian cancer cells, which have wild type p53, collected after SHetA2 treatment for 24 h. β-actin and Tom20 served as protein loading controls in cytoplasmic and mitochondrial fractions, respectively.



FIG. 7B. Mitochondrial accumulation of p53 after SHetA2 treatment in cancer cells. Bar graphs of quantified p53 band densities, which were normalized the loading controls of the western blots in FIG. 7A.



FIG. 8A. Mitochondrial accumulation of p53 after SHetA2 treatment in cancer cells. p53 levels were examined in western blots of cytoplasmic and mitochondrial fractions of OVCAR4 ovarian cancer cells, which have missense mutant p53, collected after SHetA2 treatment for 24 h. β-actin and Tom20 served as protein loading controls in cytoplasmic and mitochondrial fractions, respectively.



FIG. 8B. Mitochondrial accumulation of p53 after SHetA2 treatment in cancer cells. Bar graphs of quantified p53 band densities, which were normalized to the loading controls of the western blots in FIG. 8A.



FIG. 9A. No nuclear accumulation of p53 after SHetA2 treatment in FTSECs (non-cancerous human fallopian tube secretory epithelial cells). p53 levels were examined in western blots of cytoplasmic and mitochondrial fractions of FTSECs collected after SHetA2 treatment for 24 h. β-actin and Histone H3 served as protein loading controls in cytoplasmic and nuclear fractions, respectively.



FIG. 9B. No nuclear accumulation of p53 after SHetA2 treatment in cancer cells. Bar graphs of quantified p53 band densities, which were normalized to the loading controls in the western blots of FIG. 9A.



FIG. 10A. No mitochondrial accumulation of p53 after SHetA2 treatment in FTSECs (non-cancerous human fallopian tube secretory epithelial cells). p53 levels were examined in western blots of cytoplasmic and mitochondrial fractions of FTSECs collected after SHetA2 treatment for 24 h. β-actin and Tom20 served as protein loading controls in cytoplasmic and mitochondrial fractions, respectively.



FIG. 10B. Mitochondrial accumulation of p53 after SHetA2 treatment in cancer cells. Bar graphs of quantified p53 band densities, which were normalized to β-actin for the cytoplasmic extracts and to Tom20 for the mitochondrial extracts in the western blots of FIG. 9A.



FIG. 11. Reduction of p53 expression reduces sensitivity of cancer cells to SHetA2. Comparison of SHetA2 sensitivity in A2780 ovarian cancer cells transfected with p53 shRNA or empty vector using an MTT assay. Insert is a western blot confirming p53 reduction.



FIG. 12. Overexpression of mortalin reduces sensitivity of cancer cells to SHetA2. Sensitivity of A2780 cells transfected with a mortalin expression plasmid or empty vector were compared using a MTT assay. Insert is a western blot confirming mortalin overexpression.



FIG. 13. Ovarian cancer cells with wild type p53 are more sensitive to SHetA2 in comparison to cells that are p53 null or that express missense p53 mutants. SHetA2 sensitivities of hFTSECs and ovarian cancer cell lines with various p53 status were compared using a MTT cytotoxicity assay.



FIG. 14. Expression of missense p53 mutants increases sensitivity of SKOV3 ovarian cancer cells to PRIMA-1MET. MTT assay to compare PRIMA-1 cytotoxicity in SKOV3 parental cell line and SKOV3 sublines stably transfected with p53 mutant (R248W and R273H).



FIG. 15. PRIMA-1MET reactivates R282W mutant p53 in ovarian cancer cells. MESOV ovarian cancer cells transfected with PG13-Luc plasmid were evaluated with a luciferase reporter assay to measure the effects of SHetA2 and PRIMA-1MET on p53 transcriptional activity. ANOVA: **p ≤ 0.01.



FIG. 16. Isobologram of SHetA2 and PRIMA-1 combination in A2780 ovarian cancer cells (wild type -53). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 17. Isobologram of SHetA2 and PRIMA-1 combination in MESOV ovarian cancer cells (R282W mutant p53). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 18. Isobologram of SHetA2 and PRIMA-1 combination in SKOV3 R248W ovarian cancer cells (R248W mutant p53). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 19. Isobologram of SHetA2 and PRIMA-1 combination in Caov3 ovarian cancer cells (p53 null). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 20. Isobologram of SHetA2 and PRIMA-1 combination in non-cancerous human fallopian tube secretory epithelial cells (FTSECs). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 21. Isobologram of SHetA2 and PRIMA-1MET combination in MESOV ovarian cancer cells (R282W mutant p53). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 22. Isobologram of SHetA2 and PRIMA-1MET combination in non-cancerous human fallopian tube secretory epithelial cells (FTSECs). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 23. SHetA2, but not PRIMA-1, induces expression of the p53-regulated gene p21. qRT-PCR of mRNA isoloated from SKOV3 parental and stably-transfected p53 R273H mutant cells treated with solvent only, 20 µM PRIMA-1, 10 µM SHetA2 or the combination of 20 µM PRIMA-1 + 10 µM SHetA2. Expression values were normalized to GAPDH, and then the treated were normalized to the solvent only control samples.



FIG. 24. SHetA2, but not PRIMA-1, induces expression of the p53-regulated protein p21, while both drugs induce SLC7A11 and PARP cleavage in A2780 (wild type p53) ovarian cancer cells. Western blots of p21, SLC7A11 and PARP in the indicated cell lines after treatment with either SHetA2 or PRIMA-1 or both combined at the indicated concentrations.



FIG. 25. SHetA2, but not PRIMA-1, induces expression of the p53-regulated protein p21, while both drugs induce SLC7A11 and PARP cleavage in SKOV3 (p53 null) ovarian cancer cells. Western blots of p21, SLC7A11 and PARP in the indicated cell lines after treatment with either SHetA2 or PRIMA-1 or both combined at the indicated concentrations.



FIG. 26. SHetA2, but not PRIMA-1, induces expression of the p53-regulated protein p21, while both drugs induce SLC7A11 and PARP cleavage in SKOV3 R273H (R273H p53) ovarian cancer cells. Western blots of p21, SLC7A11 and PARP in the indicated cell lines after treatment with either SHetA2 or PRIMA-1 or both combined at the indicated concentrations.



FIG. 27. SHetA2, but not PRIMA-1, induces expression of the p53-regulated protein p21, while both drugs induce SLC7A11 and PARP cleavage in SKOV3 R248w (R248W p53) ovarian cancer cells. Western blots of p21, SLC7A11 and PARP in the indicated cell lines after treatment with either SHetA2 or PRIMA-1 or both combined at the indicated concentrations.



FIG. 28. Inhibition of caspase 3 reduces PARP-1 cleavage in A2780 (wild type p53) ovarian cancer cells. Western blots of p53, PARP-1 and caspase 3 in cultures with SHetA2 and/or PRIMA-1MET in the absence or presence of the caspase 3 inhibitor Z-DEVD-FMK.



FIG. 29. Inhibition of caspase 3 reduces PARP-1 cleavage in MES-OV (R282W p53) ovarian cancer cells. Western blots of p53, PARP-1 and caspase 3 in cultures with SHetA2 and/or PRIMA-1MET in the absence or presence of the caspase 3 inhibitor Z-DEVD-FMK.



FIG. 30. Inhibition of caspase 3 reduces PARP-1 cleavage in SKOV3 (p53 null) ovarian cancer cells. Western blots of p53, PARP-1 and caspase 3 in cultures with SHetA2 and/or PRIMA-1MET in the absence or presence of the caspase 3 inhibitor Z-DEVD-FMK.



FIG. 31. Inhibition of caspase 3 reduces PARP-1 cleavage in SKOV3 R273H (R273H p53) ovarian cancer cells. Western blots of p53, PARP-1 and caspase 3 in cultures with SHetA2 and/or PRIMA-1MET in the absence or presence of the caspase 3 inhibitor Z-DEVD-FMK.



FIG. 32. Increased sensitivity of SKOV3 cells is associated with reduced expression of SLC7A11. Bar graphs SLC7A11 band denisities normalized to the loading control (GAPDH) in FIGS. 25-27 are compared. The sensitivies of the cell lines to PRIMA-1 as shown in FIG. 14 are SKOV3 R248W>SKOV3 R273H>SKOV3 parental.



FIG. 33. Combination of SHetA2 and PRIMA-1 increases reactive oxygen species (ROS) induction in comparison the induction by either drug alone in SKOV3 (p53 null) ovarian cancer cells. ROS levels in were measured after treatment with SHetA2 or PRIMA-1 alone or in combinations **p ≤ 0.01, ***p ≤ 0.001 ****p ≤ 0.0001.



FIG. 34. Combination of SHetA2 and PRIMA-1 increases ROS induction in comparison the induction by either drug alone in SKOV3 R273H (R273H p53) ovarian cancer cells. ROS levels were measured after treatment with SHetA2 or PRIMA-1 alone or in combinations **p ≤ 0.01, ***p ≤ 0.001 ****p ≤ 0.0001.



FIG. 35. Combination of SHetA2 and PRIMA-1 increases ROS induction in comparison the induction by either drug alone in SKOV3 R248W (R248W p53) ovarian cancer cells. ROS levels were measured after treatment with SHetA2 or PRIMA-1 alone or in combinations **p ≤ 0.01, ***p ≤ 0.001 ****p ≤ 0.0001.



FIG. 36. SKOV3 ovarian cancer cell lines expressing p53 mutants have significantly higher ROS/ATP ratios in comparison to the parental p53 null line and in association with sensitivity to PRIMA-1. Bar graphs of the ratio of ROS/ATP (each normalized to protein concentration). ANOVA with Tukey’s multiple comparison test: *p <_ 0.05, **p ≤ 0.01, ****p ≤ 0.0001. The sensitivities of the cell lines to PRIMA-1 as shown in FIG. 14 are SKOV3 R248W>SKOV3 R273H>SKOV3 parental p53 null cells.



FIG. 37. Combination of SHetA2 and PRIMA-1MET significantly increases the ROS/ATP ratio over the elevated ratio caused by either drug alone in A2780 (wild type p53) ovarian cancer cells. Ratio of ROS/ATP in A2780 cells after treatment with SHetA2 or PRIMA-1MET or both combined.



FIG. 38. Reduction of p53 significantly decreased sensitivity of A2780 (wild type p53) ovarian cancer cells to ATP depletion caused by SHetA2 and PRIMA-1MET combination treatment. ATP concentrations were measured in A2780 cells stably transfected with control shRNA or p53 shRNA after treatment with SHetA2 and PRIMA-1MET drug combination.



FIG. 39. SHetA2 or PRIMA-1MET had no significant effects on body weight or ascites (measured by abdominal size) in nu/nu immunocompromised mice injected intraperitoneally with MESOV3 ovarian cancer cells. Mice body weights were measured during the duration of treatment duration and abdominal size was measured at the end of the treatment period. ANOVA: ** p<0.01 for the untreaed control weight compared to the other groups (likely due to cancer-induced cachexia).



FIG. 40. Fewer tumors developed in intraperitoneally MESOV ovarian cancer cell-injected mice treated with SHetA2 and/or PRIMA-1MET. Photos of peritoneal tumors harvested from MESOV ovarian cancer cell - injected athymic nude mice after the treatment period.



FIG. 41. SHetA2 and PRIMA-1MET significantly increased in an additive manner the tumor free rates in nu/nu mice injected intraperitoneally with MESOV ovarian cancer cells. Tumor free rates in the various treatment groups, Linear Regression Model: SHetA2 (p=0.004, OR=10.384, 95% CI: 2.158, 48.965) and PRIMA1MET (p=0.048, OR=4.464, 95% CI: 1.014, 19.655) functioned additively in preventing tumor development.



FIG. 42. SHetA2 or PRIMA-1MET did not cause kidney or liver toxicity in nu/nu mice injected intraperitoneally with MESOV ovarian cancer cells. 20X images of H & E staining of liver and kidney specimen of mice tumor model to determine toxicity of drug combination. The kidneys exhibited glomeruli that were well formed without inflammation and the tubules were normal with no necrosis, apoptosis or inflammation. Livers exhibited no necrosis, fat deposition or inflammation.



FIG. 43A. Combination of SHetA2 and PRIMA-1MET significantly increased SLC7A11 in tumors of nu/nu mice injected intraperitoneally with MESOV ovarian cancer cells. Western blot analysis was performed on tumors from three randomly chosen mice in each treatment group to analyze expression of Pax8 (marker of ovarian cancer), SLC7A11 (marker of PRIMA-1/PRIMA-1MET sensitivity) and p53 in mice tumor specimens.



FIG. 43B. Combination of SHetA2 and PRIMA-1MET significantly increased SLC7A11 in tumors of nu/nu mice injected intraperitoneally with MESOV ovarian cancer cells. Bar graph of p53 and SLC7A11 band densities normalized to the loading controls in the western blot of FIG. 43A.. ANOVA: *p<0.05



FIG. 44. Isobologram of SHetA2 and palbociclib combination in SiHa cervical cancer cells (human papillomavirus positive). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 45. Isobologram of SHetA2 and palbociclib combination in Caski cervical cancer cells (human papillomavirus positive). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 46. Isobologram of SHetA2 and palbociclib combination in C33a cervical cancer cells (human papillomavirus negative). Fold effects of a 2-fold dilution series of a drug combination mixed at 1:1 ratio of the EC50s of each drug were compared with single drug dilution series using the Chou-Talaly method.



FIG. 47. Cell Death Induction by SHetA2 and Palbociclib. Photomicrographs of SiHa cultures treated for 24 hours with each drug alone and in combination as indicated. Rounding up of the cells indicates cell death.



FIG. 48. Cell Death Induction by SHetA2 and Palbociclib. Photomicrographs of C33A cultures treated for 24 hours with each drug alone and in combination as indicated. Rounding up of the cells indicates cell death.



FIG. 49. SHetA2 and palbociclib significantly reduce SiHa cervical cancer cell line xenograft tumor growth in an additive manner. Immunocompromized mice bearing SiHa xenografts were gavaged with 60 mg/kg SHetA2, 100 mg/kg palbociclib or the combination every other day. Controls were gavaged with the same Kolliphor HS15 used to suspend the drugs. Using a generalized estimating equations (GEE) model to analyze tumor growth treated as a continuous variable, there was a significant additive effect of SHetA2 (p<0.001) and palbociclib (p<0.001) on day 65 of treatment. This additive effect between SHetA2 (p=0.001) and palbociclib (p=0.051) was trending significant at day 63 of treatment. Additionally, SHetA2 starts to show significant tumor suppressing activity as early as day 63 of treatment (p=0.045).



FIG. 50 shows synergy between SHetA2 and palbociclib in a combination treatment of the human endometrial cancer cell line Hec1B. Cells were treated with a dilution series of palbociclib, SHetA2, or a 1:1 ratio of the half maximal inhibitory concentrations of palbociclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa). Circles: Fa=0.5, squares: Fa=0.75, triangles: Fa=0.9. Y axis = Palbociclib dose (µM).



FIG. 51 shows synergy between SHetA2 and palbociclib in a combination treatment of the human endometrial cancer cell line Ishikawa. Cells were treated with a dilution series of palbociclib, SHetA2, or a 1:1 ratio of the half maximal inhibitory concentrations of palbociclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa). Circles: Fa=0.5, squares: Fa=0.75, triangles: Fa=0.9. Y axis = Palbociclib dose (µM).



FIG. 52 shows results of treating three ovarian cancer cell lines (OVCAR-8, OV90, and ES2) with a combination treatment of SHetA2 (5 µM) and abemaciclib (2 µM). The three ovarian cancer cell lines treated with the drug combination exhibited significantly greater growth inhibition in comparison to single drug treatments after 48 hr of treatment. ** ANOVA p values <0.01, ***:ANOVA p values <0.001, ****: ANOVA P values < 0.0001.



FIG. 53 shows synergy between SHetA2 and abemaciclib in the human ovarian cancer cell line MESOV. The indicated cell lines were treated with a dilution series of abemaciclib, SHetA2 or 1:1 ratio of the half maximal inhibitory concentrations of abemaciclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa). Circles: Fa=0.5, squares: Fa=0.75, triangles: Fa=0.9. Y axis = Abemaciclib dose (µM).



FIG. 54 shows synergy between SHetA2 and abemaciclib in the human ovarian cancer cell line. OV90. The indicated cell lines were treated with a dilution series of abemaciclib, SHetA2 or 1:1 ratio of the half maximal inhibitory concentrations of abemaciclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa). Circles: Fa=0.5, squares: Fa=0.75, triangles: Fa=0.9. Y axis = Abemaciclib dose (µM).



FIG. 55 shows synergy between SHetA2 and abemaciclib in the human endometrial cancer cell line AN3CA. The indicated cell lines were treated with a dilution series of abemaciclib, SHetA2 or 1:1 ratio of the half maximal inhibitory concentrations of abemaciclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa). Circles: Fa=0.5, squares: Fa=0.75, triangles: Fa=0.9. Y axis = Abemaciclib dose (µM).



FIG. 56 shows synergy between SHetA2 and abemaciclib in the human endometrial cancer cell line Heclb. The indicated cell lines were treated with a dilution series of abemaciclib, SHetA2 or 1:1 ratio of the half maximal inhibitory concentrations of abemaciclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa). Circles: Fa=0.5, squares: Fa=0.75, triangles: Fa=0.9. Y axis = Abemaciclib dose (µM).



FIG. 57 shows synergy between SHetA2 and abemaciclib in the human endometrial cancer cell line Ishikawa. The indicated cell lines were treated with a dilution series of abemaciclib, SHetA2 or 1:1 ratio of the half maximal inhibitory concentrations of abemaciclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa). Circles: Fa=0.5, squares: Fa=0.75, triangles: Fa=0.9. Y axis = Abemaciclib dose (µM).



FIG. 58 shows results of treating three ovarian cancer cell lines (OVCAR-8, OV90, and ES2) with a combination treatment of SHetA2 (5 µM) and ON123300 (7.5 µM). The three ovarian cancer cell lines treated with the drug combination exhibited significantly greater growth inhibition in comparison to single drug treatments on after 24 hr of treatment. ** ANOVA p values <0.01, ***:ANOVA p values <0.001, ****: ANOVA P values < 0.0001.





DETAILED DESCRIPTION

Many cancer deaths, including most deaths due to ovarian cancer, are caused by disease recurrence. Current maintenance therapies to reduce recurrence of cancer are often limited by toxicity and no proven effect on overall survival. The present disclosure is directed to synergistically-effective drug combinations comprising a heteroarotinoid (e.g., SHetA2), and at least one of an Azabicyclooctan-3-one derivative (e.g., PRIMA-1 or PRIMAMET), and a CDK4/6 inhibitor (e.g., Palbociclib, Abemaciclib, Ribociclib, Narazaciclib, Dalpiciclib, Dinaciclib, Milciclib, or Seliciclib), which have a synergistic effect in causing cancer cell death, reduction of tumor size, reduction of tumor growth, inhibition of cancer metastases, and/or inhibition of tumor recurrence. The present work demonstrated, for example, that SHetA2, a mortalin-targeting drug, and PRIMA-1MET, a p53 reactivator drug, inhibit ovarian cancer cell line growth synergistically, thus can be used in combination for ovarian cancer treatment and/or maintenance therapy (to prevent recurrence), as well as for treatment and/or maintenance therapy of other cancers.


Before further detailed description of various embodiments of the compositions and methods of use thereof of the present disclosure, it is to be understood that the present disclosure is not limited in application to the details of methods and compositions as set forth in the following description. The description provided herein is intended for purposes of illustration only and is not intended to be construed in a limiting sense. The present disclosure is capable of other embodiments or of being practiced or carried out in various ways. As such, the language used herein is intended to be given the broadest possible scope and meaning; and the embodiments are meant to be exemplary, not exhaustive. Also, it is to be understood that the phraseology and terminology employed herein is for the purpose of description and should not be regarded as limiting unless otherwise indicated as so. Moreover, in the following detailed description, numerous specific details are set forth in order to provide a more thorough understanding of the disclosure. However, it will be apparent to a person having ordinary skill in the art that various embodiments of the present disclosure may be practiced without these specific details. In other instances, features which are well known to persons of ordinary skill in the art have not been described in detail to avoid unnecessary complication of the description. It is intended that all alternatives, substitutions, modifications and equivalents apparent to those having ordinary skill in the art are included within the scope of the present disclosure as defined herein. Thus the examples described below, which include particular embodiments, will serve to illustrate the practice of the present disclosure, it being understood that the particulars shown are by way of example and for purposes of illustrative discussion of particular embodiments only and are presented in the cause of providing what is believed to be a useful and readily understood description of procedures as well as of the principles and conceptual aspects of the inventive concepts. Thus, while the compositions and methods of the present disclosure have been described in terms of particular embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit, and scope of the inventive concepts disclosed herein.










List of Abbreviations




CDK4/6:
cyclin-dependent kinase 4 and 6,


CI:
Combination index,


DRI:
Dose-reduction index,


FDA:
Food and Drug Administration,


GOG:
Gynecologic Oncology Group,


HGSOC:
High-grade serous ovarian cancer,


hFTSECs:
Human fallopian tube Secretory epithelial cells,


IC:
Inhibitory concentration,


IC50:
Half maximal inhibitory concentration,


M-p53:
Missense mutant p53,


MTT:
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide,


P53:
Tumor protein 53,


PARP:
Poly ADP ribose polymerase,


PRIMA-1:
2,2-Bishydroxymethyl-1-azabicyclo[2.2.2]octan-3-one,


PRIMA-1MET:
2-(hydroxymethyl)-2-(methoxymethyl)-1-azabicyclo [2.2.2]octan-3-one,


RAID:
Rapid Access to Intervention Development,


RAPID:
Rapid Access to Preventive Intervention Development,


SHetA2:
[(4-nitrophenyl)amino][2,2,4,4-tetramethylthiochroman-6-yl]amino] methane-thione,


TCGA:
The Cancer Genome Atlas,


TMA:
Tumor Microarray,


TP53:
Tumor protein 53 gene,


OS:
Overall Survival,


ROS:
Reactive oxygen species.






Each patent, published patent application, and non-patent publication referenced in any portion of this application, including but not limited to U.S. Ser. No. 18/145,334, filed Dec. 22, 2022, U.S. Ser. No. 17/276,032, filed Mar. 12, 2021, International Application No. PCT/US2019/050775, filed Sept. 12, 2019, and U.S. Serial No. 62/730,345, filed Sept. 12, 2018, is expressly incorporated herein by reference in its entirety to the same extent as if the individual patent, or published patent application, or non-patent publication was specifically and individually indicated to be incorporated by reference.


Unless otherwise defined herein, scientific and technical terms used in connection with the present disclosure shall have the meanings that are commonly understood by those having ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.


As utilized in accordance with the methods and compositions of the present disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings:


The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.” The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or when the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and “and/or.” The use of the term “at least one” will be understood to include one as well as any quantity more than one, including but not limited to, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 100, or any integer inclusive therein. The term “at least one” may extend up to 100 or 1000 or more, depending on the term to which it is attached; in addition, the quantities of 100/1000 are not to be considered limiting, as higher limits may also produce satisfactory results. In addition, the use of the term “at least one of X, Y and Z” will be understood to include X alone, Y alone, and Z alone, as well as any combination of X, Y and Z.


As used herein, all numerical values or ranges include fractions of the values and integers within such ranges and fractions of the integers within such ranges unless the context clearly indicates otherwise. Thus, to illustrate, reference to a numerical range, such as 1-10 includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, as well as 1.1, 1.2, 1.3, 1.4, 1.5, etc., and so forth. Reference to a range of 1-50 therefore includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, etc., up to and including 50, as well as 1.1, 1.2, 1.3, 1.4, 1.5, etc., 2.1, 2.2, 2.3, 2.4, 2.5, etc., and so forth. Reference to a series of ranges includes ranges which combine the values of the boundaries of different ranges within the series. Thus, to illustrate reference to a series of ranges, for example, of 1-10, 10-20, 20-30, 30-40, 40-50, 50-60, 60-75, 75-100, 100-150, 150-200, 200-250, 250-300, 300-400, 400-500, 500-750, 750-1,000, includes ranges of 1-20, 10-50, 50-100, 100-500, and 500-1,000, for example.


As noted above, any numerical range listed or described herein is intended to include, implicitly or explicitly, any number or sub-range within the range, particularly all integers, including the end points, and is to be considered as having been so stated. For example, “a range from 1.0 to 10.0” is to be read as indicating each possible number, including integers and fractions, along the continuum between and including 1.0 and 10.0, that is, having a minimum value equal to or greater than 1.0 and a maximum value equal to or less than 10.0, such as, for example, 3.25 to 8.65. Any maximum numerical limitation recited herein is intended to include all lower numerical limitations subsumed therein, and any minimum numerical limitation recited in this specification is intended to include all higher numerical limitations subsumed therein. Accordingly, Applicant reserves the right to amend this specification, including the claims, to expressly recite any sub-range subsumed within the ranges expressly recited herein. Thus, even if a particular data point within the range is not explicitly identified or specifically referred to, it is to be understood that any data points within the range are to be considered to have been specified, and that the inventor(s) possessed knowledge of the entire range and the points within the range.


As used in this specification and claims, the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or openended and do not exclude additional, unrecited elements or method steps.


The term “or combinations thereof” as used herein refers to all permutations and combinations of the listed items preceding the term. For example, “A, B, C, or combinations thereof” is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AAB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.


Throughout this application, the terms “about” or “approximately” are used to indicate that a value includes the inherent variation of error for the composition, the method used to administer the composition, or the variation that exists among the study subjects. As used herein the qualifiers “about” or “approximately” are intended to include not only the exact value, amount, degree, orientation, or other qualified characteristic or value, but are intended to include some slight variations due to measuring error, manufacturing tolerances, observer error, and combinations thereof, for example. The term “about” or “approximately”, where used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass, for example, variations of ± 20% or ± 10%, or ± 5%, or ± 1%, or ± 0.1% from the specified value, as such variations are appropriate to perform the disclosed methods and as understood by persons having ordinary skill in the art. As used herein, the term “substantially” means that the subsequently described event or circumstance completely occurs or that the subsequently described event or circumstance occurs to a great extent or degree. For example, the term “substantially” means that the subsequently described event or circumstance occurs at least 80% of the time, at least 90% of the time, at least 91% of the time, at least 92% of the time, at least 93% of the time, at least 94% of the time, at least 95% of the time, at least 96% of the time, at least 97% of the time, at least 98% of the time, or at least 99% of the time.


As used herein any reference to “one embodiment” or “an embodiment” means that a particular element, feature, composition, structure, or characteristic described in connection with the embodiment is included in at least one embodiment. The appearances of the phrase “in one embodiment” in various places in the specification are not necessarily all referring to the same embodiment.


The term “mutant” or “variant” is intended to refer to a protein, peptide, nucleic acid or organism which has at least one amino acid or nucleotide which is different from the wild type version of the protein, peptide, nucleic acid, or organism and includes, but is not limited to, point substitutions, multiple contiguous or non-contiguous substitutions, chimeras, or fusion proteins, and the nucleic acids which encode them. Examples of conservative amino acid substitutions include, but are not limited to, substitutions made within the same group such as within the group of basic amino acids (such as arginine, lysine, histidine), acidic amino acids (such as glutamic acid and aspartic acid), polar amino acids (such as glutamine and asparagine), hydrophobic amino acids (such as leucine, isoleucine, and valine), aromatic amino acids (such as phenylalanine, tryptophan, tyrosine) and small amino acids (such as glycine, alanine, serine, threonine, methionine). Other examples of possible substitutions are described below.


The term “pharmaceutically acceptable” refers to compounds and compositions which are suitable for administration to humans and/or animals without undue adverse side effects such as toxicity, irritation and/or allergic response commensurate with a reasonable benefit/risk ratio.


As used herein, “pure,” or “substantially pure” means an object species is the predominant species present (i.e., on a molar basis it is more abundant than any other object species in the composition thereof), and particularly a substantially purified fraction is a composition wherein the object species comprises at least about 50 percent (on a molar basis) of all macromolecular species present. Generally, a substantially pure composition will comprise more than about 80% of all macromolecular species present in the composition, more particularly more than about 85%, more than about 90%, more than about 95%, or more than about 99%. The term “pure” or “substantially pure” also refers to preparations where the object species (e.g., the active agent) is at least 60% (w/w) pure, or at least 70% (w/w) pure, or at least 75% (w/w) pure, or at least 80% (w/w) pure, or at least 85% (w/w) pure, or at least 90% (w/w) pure, or at least 92% (w/w) pure, or at least 95% (w/w) pure, or at least 96% (w/w) pure, or at least 97% (w/w) pure, or at least 98% (w/w) pure, or at least 99% (w/w) pure, or 100% (w/w) pure.


The terms “subject” and “patient” are used interchangeably herein and will be understood to refer to a warm blooded animal, particularly a mammal. Non-limiting examples of animals within the scope and meaning of this term include dogs, cats, rabbits, rats, mice, guinea pigs, chinchillas, hamsters, ferrets, horses, pigs, goats, cattle, sheep, zoo animals, camels, llamas, non-human primates, including Old and New World monkeys and non-human primates (e.g., cynomolgus macaques, chimpanzees, rhesus monkeys, orangutans, and baboons), and humans.


The term “active agent” where used herein is intended to refer to a substance which possesses a biological activity relevant to the present disclosure, and particularly refers to therapeutic and diagnostic substances which may be used in methods described in the present disclosure. By “biologically active” is meant the ability to modify the physiological system of a cell, tissue, or organism without reference to how the active agent has its physiological effects. Where used herein, unless otherwise noted, the term “active agent” includes pharmaceutically-acceptable salts, hydrates, solvates, and amorphous solids thereof.


Therapeutic drug combinations of the present disclosure include, but are not limited to, a first active agent selected from heteroarotinoids, particularly flexible heteroarotinoids such as arefurther described below; and a second active agent selected from the group consisting of Azabicyclooctan-3-one derivatives, and cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors.


Non-limiting examples of Azabicyclooctan-3-one derivatives include PRIMA-1 (NSC-281668), PRIMA-1MET (Eprenetapopt, APR246), and analogs thereof shown in International Patent Publications WO2002/024692 and WO2003/070250, and U.S. Pat. 7,759,361. Where used herein “PRIMA-1/PRIMA-1MET” refers to either PRIMA-1 or PRIMA-1MET.


Particular examples of heteroarotinoids include flexible heteroarotinoids having a urea or thiourea linker (referred to as “Flex-Hets”), such as, but not limited to any heteroarotinoid disclosed in U.S. Pat. Nos. 6,586,460 (see, for example, Columns 2-5 thereof) and 7,612,107 (see, for example, Columns 7-9 thereof). Non-limiting examples of heteroarotinoids that may be used herein include SHetA2, SHetA3, SHetA4, SHetC2, SHetD3, SHetD4, SHetD5, SHet50, SHet65, SHet100, OHet72, NHet17, NHet86, NHet90, and other heteroarotinoid compounds shown in Table 1, compounds 1-3 shown in Table 2, heteroarotinoid compounds shown in Table 3, heteroarotinoid compounds shown in Table 4, heteroarotinoid compounds shown in Table 5, heteroarotinoid compounds shown in Table 6, heteroarotinoid compounds shown in Table 7, and heteroarotinoid compounds shown in Table 8.





TABLE 1







Heteroarotinoids (Benbrook et al., 1997*)






embedded image


56


embedded image


43




embedded image


54


embedded image


40




embedded image


52


embedded image


35.7




embedded image


51


embedded image


35.8




embedded image


50


embedded image


33




embedded image


50


embedded image


31




embedded image


46


embedded image


29


*D. M. Benbrook et al., “Biologically Active Heteroarotinoids Exhibiting Anticancer Activity and Decreased Toxicity,” J. Med. Chem., 1977, 40 (3567-3583).









TABLE 2






Nitrogen Heteroarotinoids (Dhar et al., 1999*)






embedded image




embedded image




embedded image




*A. Dhar et al., “Synthesis, Structure-Activity Relationships, and RARy-Ligand Interactions of Nitrogen Heteroarotinoids,” J. Med. Chem., 1999, 42 (3602-3614).









TABLE 3






Heteroarotinoids (Zacheis et al., 1997*)






embedded image




embedded image




embedded image






embedded image




embedded image




embedded image






embedded image




embedded image




embedded image






embedded image




embedded image




embedded image






embedded image




embedded image




embedded image




*D. Zacheis et al., “Heteroarotinoids Inhibit Head and Neck Cancer Cell Lines in Vitro and in Vivo Through Both RAR and RXR Retinoic Acid Receptors,” J. Med. Chem., 1999, 42 (4434-4445).









TABLE 4







Heteroarotinoids (Brown et al., 2004*)






embedded image


0.5-1.0


embedded image


2.0-4.0




embedded image


5.0-10.0


embedded image


10.0-20.0




embedded image


20.0-40.0


embedded image


20.0-40.0




embedded image


20.0-40.0


embedded image


20.0-40.0




embedded image


20.0-40.0


embedded image


20.0-40.0




embedded image


20.0-40.0


embedded image


20.0-40.0




embedded image


20.0-40.0


embedded image


20.0-40.0




embedded image


20.0-40.0


embedded image


20.0-40.0


*C. W. Brown et al., “Novel Heteroarotinoids as Potential Antagonists of Mycobacterium bovis BCG,” J. Med. Chem., 2004, 47 (1008-1017).









TABLE 5





Heteroarotinoids (Le et al., 2007*)






embedded image




embedded image




9 a, X = S; Z = NO2
10 a. X = S; Z = NO2


b. X = O; Z = NO2
b. X = S; Z = CO2Et


c. X = O; Z = CO2Et
c. X = O; Z = NO2



d. X = O; Z = CO2Et


*T.C. Le et al., “Heteroarotinoids with Anti-Cancer Activity Against Ovarian Cancer Cells,” The Open Medicinal Chemistry Journal, 2007, 1 (11-23).









TABLE 6










Heteroarotinoids (Liu et al., 2004*)






embedded image - 15a-h















entry
R
X
R
Z
product
yield (%)




1
H
O
H
CO2-Et
15a
64


2
H
S
H
CO2Et
15b
92


3
H
S
H
NO2
15c
83


4
H
S
H
SO2NH2
15d
87


5
H
S
CH3
NO2
15e
82


6
CH3
O
H
Co2Et
15f
43


7
CH3
S
H
CO2Et
15g
66


8
CH3
S
H
SO2NH2
15h
56


*S. Liu et al., “Synthesis of Flexible Sulfur-Containing Heteroarotinoids That Induce Apoptosis and Reactive Oxygen Species with Discrimination between Malignant and Benign Cells,” J. Med. Chem., 2004, 47 (999-1007).









TABLE 7





Heteroarotinoids (Gnanasekaran et al., 2015*)




embedded image




Compound
Y


Nitrogen-Substituted Flex-Hets




5 (SHetA2)
4-NO2


18b
4-NO2,


18f
4-N(CH3)2


18h
4-NH2


20a
4-N-methylamide


20b
4-N-cyclopropylamide


20c
4-oxomorpholino








Non-Nitrogen-Substituted Flex-Hets




18a
4-CO2Et


18c
4-CF3


18d
2,3,4,5,6-F


18e
3,4-OCH3


18g
4-CO2H


*K. K. Gnanasekaran et al., “Synthesis and evaluation of second generation Flex-Het scaffolds against the human ovarian A2780 cancer cell line,” European J. Med. Chem., 2015, 96 (209-217).









TABLE 8




Heteroarotinoids (Liu et al., 2009*)






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image




*T. Liu et al., “Development of flexible-heteroarotinoids for kidney cancer,” Mol. Cancer Ther., 2009, 8(5), (1227-1238).






Particular CDK4/6 inhibitors that may be used in the methods and compositions of the present disclosure include, but are not limited to, Palbociclib (PD0332991, Ibrance®), Abemaciclib (LY28352191, Verzenio®), Ribociclib (LEE011, Kisqali®), Narazaciclib (ON123300), Dalpiciclib (SRH6390), Dinaciclib (SCH727965), Milciclib (PHA-848125) and Seliciclib (Roscovitine, CYC202). Other CDK4/6 inhibitors that may be used in the present disclosure include, but are not limited to, the compounds labeled herein as Wang-4d and Wang-4e in Table 9 herein, CDK4/6 inhibitory compounds in U.S. Published Pat. Application No. 2014/0227222, compounds represented by Formulas I-V and Table 1 in U.S. Published Pat. Application No. 2017/0246171, and compounds represented by Formula II of U.S. Published Pat. Application No. 2019/0209566.





TABLE 9




CDK4/6 inhibitors (P. Wang et al., 2016*)




Wang-4d:




embedded image




Wang-4e:




embedded image




*P. Wang, et al., “New palbociclib analogues modifies at the terminal piperazine ring and their anticancer activities,” Euro. J. of Med. Chem., 2016, 122, (546-556).






Where used herein the terms heteroarotinoid, Azabicyclooctan-3-one derivative, and CDK4/6 inhibitor, and specific examples thereof, are intended to include pharmaceutically-acceptable salts, hydrates, solvates, and amorphous solids thereof.


Particular therapeutic drug combinations of the present disclosure include, but are not limited to, a first active agent selected from at least one heteroarotinoid, and at least one Azabicyclooctan-3-one derivative. Examples of such drug combinations are shown in Table 10.





TABLE 10






Heteroarotinoid and PRIMA-1 /PRIMA-1MET Drug Combinations



PRIMA-1
PRIMA-1MET




SHetA2
SHetA2+ PRIMA-1
SHetA2+ PRIMA-1MET


SHetA3
SHetA3+ PRIMA-1
SHetA3 +PRIMA-1MET


SHetA4
SHetA4+ PRIMA-1
SHetA4 +PRIMA-1MET


SHetC2
SHetC2+ PRIMA-1
SHetC2 +PRIMA-1MET


SHetD3
SHetD3+ PRIMA-1
SHetD3 +PRIMA-1MET


SHetD4
SHetD4+ PRIMA-1
SHetD4 +PRIMA-1MET


SHetD5
SHetD5+ PRIMA-1
SHetD5 +PRIMA-1MET


SHet50
SHet50+ PRIMA-1
SHet50 +PRIMA-1MET


SHet65
SHet65+ PRIMA-1
SHet65 +PRIMA-1MET


SHet100
SHet100+ PRIMA-1
SHet100 +PRIMA-1MET


OHet72
OHet72+ PRIMA-1
OHet72 +PRIMA-1MET


NHet17
NHetl7+ PRIMA-1
NHet17 +PRIMA-1MET


NHet86
NHet86+ PRIMA-1
NHet86 +PRIMA-1MET


NHet90
NHet90+ PRIMA-1
NHet90 +PRIMA-1MET






Particular therapeutic drug combinations of the present disclosure also include, but are not limited to, a first active agent selected from at least one heteroarotinoid, and at least one CDK4/6 inhibitor. Examples of such drug combinations are shown in Tables 11, 12, and 13.





TABLE 11








Heteroarotinoid and CDK4/6 Inhibitor Drug Combinations



Palbociclib (PBC)
Abemaciclib(AMC)
Ribociclib(RBC)
ON123300 (ON1)




SHetA2
SHetA2+PBC
SHetA2+AMC
SHetA2+RBC
SHetA2+ON1


SHetA3
SHetA3+PBC
SHetA3+AMC
SHetA3+RBC
SHetA3+ON1


SHetA4
SHetA4+PBC
SHetA4+AMC
SHetA4+RBC
SHetA4+ON1


SHetC2
SHetC2+PBC
SHetC2+AMC
SHetC2+RBC
SHetC2+ON1


SHetD3
SHetD3+PBC
SHetD3+AMC
SHetD3+RBC
SHetD3+ON1


SHetD4
SHetD4+PBC
SHetD4+AMC
SHetD4+RBC
SHetD4+ON1


SHetD5
SHetD5+PBC
SHetD5+AMC
SHetD5+RBC
SHetD5+ON1


SHet50
SHet50+PBC
SHet50+AMC
SHet50+RBC
SHet50+ON1


SHet65
SHet65+PBC
SHet65+AMC
SHet65+RBC
SHet65+ON1


SHet100
SHet100+PBC
SHet100+AMC
SHet100+RBC
SHet100+ON1


OHet72
OHet72+PBC
OHet72+AMC
OHet72+RBC
OHet72+ON1


NHet17
NHet17+PBC
NHet17+AMC
NHet17+RBC
NHet17+ON1


NHet86
NHet86+PBC
NHet86+AMC
NHet86+RBC
NHet86+ON1


NHet90
NHet90+PBC
NHet90+AMC
NHet90+RBC
NHet90+ON1









TABLE 12








Heteroarotinoid and CDK4/6 Inhibitor Drug Combinations



Dalpiciclib (DPC)
Dinaciclib (DNC)
Milciclib (MLC)
Seliciclib (SLC)




SHetA2
SHetA2+DPC
SHetA2+DNC
SHetA2+MLC
SHetA2+SLC


SHetA3
SHetA3+DPC
SHetA3+DNC
SHetA3+MLC
SHetA3+SLC


SHetA4
SHetA4+DPC
SHetA4+DNC
SHetA4+MLC
SHetA4+SLC


SHetC2
SHetC2+DPC
SHetC2+DNC
SHetC2+MLC
SHetC2+SLC


SHetD3
SHetD3+DPC
SHetD3+DNC
SHetD3+MLC
SHetD3+SLC


SHetD4
SHetD4+DPC
SHetD4+DNC
SHetD4+MLC
SHetD4+SLC


SHetD5
SHetD5+DPC
SHetD5+DNC
SHetD5+MLC
SHetD5+SLC


SHet50
SHet50+DPC
SHet50+DNC
SHet50+MLC
SHet50+SLC


SHet65
SHet65+DPC
SHet65+DNC
SHet65+MLC
SHet65+SLC


SHet100
SHet100+DPC
SHet100+DNC
SHet100+MLC
SHet100+SLC


OHet72
OHet72+DPC
OHet72+DNC
OHet72+MLC
OHet72+SLC


NHet17
NHet17+DPC
NHet17+DNC
NHet17+MLC
NHet17+SLC


NHet86
NHet86+DPC
NHet86+DNC
NHet86+MLC
NHet86+SLC


NHet90
NHet90+DPC
NHet90+DNC
NHet90+MLC
NHet90+SLC









TABLE 13






Heteroarotinoid and CDK4/6 Inhibitor Drug Combinations



Wang-4d (W4d)
Wang-4e (W4e)




SHetA2
SHetA2+W4d
SHetA2+W4e


SHetA3
SHetA3+W4d
SHetA3+W4e


SHetA4
SHetA4+W4d
SHetA4+W4e


SHetC2
SHetC2+W4d
SHetC2+W4e


SHetD3
SHetD3+W4d
SHetD3+W4e


SHetD4
SHetD4+W4d
SHetD4+W4e


SHetD5
SHetD5+W4d
SHetD5+W4e


SHet50
SHet50+W4d
SHet50+W4e


SHet65
SHet65+W4d
SHet65+W4e


SHet100
SHet100+W4d
SHet100+W4e


OHet72
OHet72+W4d
OHet72+W4e


NHet17
NHet17+W4d
NHet17+W4e


NHet86
NHet86+W4d
NHet86+W4e


NHet90
NHet90+ W 4d
NHet90+W4e






“Treatment” refers to therapeutic treatments. “Prevention” refers to prophylactic or preventative treatment measures or reducing the onset of a condition or disease. The term “treating” refers to administering the composition to a subject for therapeutic purposes and/or for prevention. Non-limiting examples of modes of administration include inhalation, oral, topical, retrobulbar, subconjunctival, transdermal, parenteral, subcutaneous, intranasal, intramuscular, intraperitoneal, intravitreal, and intravenous routes, including both local and systemic applications. In addition, the compositions of the present disclosure may be designed to provide delayed, controlled, extended, and/or sustained release using formulation techniques which are well known in the art.


The terms “therapeutic composition” and “pharmaceutical composition” refer to an active agent-containing composition that may be administered to a subject by any method known in the art or otherwise contemplated herein, wherein administration of the composition brings about a therapeutic effect as described elsewhere herein. In addition, the compositions of the present disclosure may be designed to provide delayed, controlled, extended, and/or sustained release using formulation techniques which are well known in the art.


Where used herein the term “drug combination” refers to a combination of drugs (compounds) which can be conjointly administered.


As used herein the terms “conjoint,” “conjointly,” “conjointly administered,” or “conjoint administration” refers to any form of administration of a combination of two or more different therapeutic compounds (also referred to herein as drugs or active agents) such that the second compound is administered while the previously administered therapeutic compound is still effective in the body, whereby the two or more compounds are simultaneously active in the patient, enabling a synergistic interaction of the compounds. For example, the different therapeutic compounds can be administered either together in the same formulation (i.e., as a physical mixture), or in separate formulations, either concomitantly (at the same time) or sequentially. When administered sequentially the different compounds may be administered immediately in succession, or separated by a suitable duration of time, as long as the active agents function together in a synergistic manner.


The term “effective amount” refers to an amount of an active agent which is sufficient to exhibit a detectable therapeutic effect without excessive adverse side effects (such as toxicity, irritation and allergic response) commensurate with a reasonable benefit/risk ratio when used in the manner disclosed herein. The effective amount for a patient will depend upon the type of patient, the patient’s size and health, the nature and severity of the condition to be treated, the method of administration, the duration of treatment, the nature of concurrent therapy (if any), the specific formulations employed, and the like. Thus, it is not possible to specify an exact effective amount in advance. However, the effective amount for a given situation can be determined by one of ordinary skill in the art using routine experimentation based on the information provided herein.


The term “ameliorate” means a detectable or measurable improvement in a subject’s condition, disease or symptom thereof. A detectable or measurable improvement includes a subjective or objective decrease, reduction, inhibition, suppression, limit or control in the occurrence, frequency, severity, progression, or duration of the condition or disease, or an improvement in a symptom or an underlying cause or a consequence of the disease, or a reversal of the disease. A successful treatment outcome can lead to a “therapeutic effect,” or “benefit” of ameliorating, decreasing, reducing, inhibiting, suppressing, limiting, controlling or preventing the occurrence, frequency, severity, progression, or duration of a disease or condition, or consequences of the disease or condition in a subject.


A decrease or reduction in worsening, such as stabilizing the condition or disease, is also a successful treatment outcome. A therapeutic benefit therefore need not be complete ablation or reversal of the disease or condition, or any one, most or all adverse symptoms, complications, consequences or underlying causes associated with the disease or condition. Thus, a satisfactory endpoint may be achieved when there is an incremental improvement such as a partial decrease, reduction, inhibition, suppression, limit, control, or prevention in the occurrence, frequency, severity, progression, or duration, or inhibition or reversal of the condition or disease (e.g., stabilizing), over a short or long duration of time (hours, days, weeks, months, etc.). Effectiveness of a method or use, such as a treatment that provides a potential therapeutic benefit or improvement of a condition or disease, can be ascertained by various methods and testing assays.


Where used herein the term cancer, cancer cell, or tumor, can be used in reference to various cancers including, but not limited to ovarian cancer, cervical cancer, peritoneal cancer, uterine cancer, vulvar cancer, oral cancer, pharyngeal cancer, oropharyngeal cancer, nasopharyngeal cancer, respiratory cancer, urogenital cancer, gastrointestinal cancer, central or peripheral nervous system tissue cancer, an endocrine cancer, neuroendocrine cancer, glioma, carcinoma, lymphoma, melanoma, fibroma, meningioma, brain cancer, renal cancer, pheochromocytoma, pancreatic islet cell cancer, Li-Fraumeni tumors, thyroid cancer, parathyroid cancer, pituitary tumors, adrenal gland tumors, osteogenic sarcoma tumors, multiple neuroendocrine type I and type II tumors, head and neck cancer, prostate cancer, esophageal cancer, tracheal cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, uterine cancer, testicular cancer, colon cancer, rectal cancer, and skin cancer, and any other cancer which comprises one or more mutations in TP53 gene or which is driven by cyclin D1/cdk4/6 complex.


In other embodiments, the term cancer, cancer cell, or tumor, can be used in reference to various cancers including, but not limited to breast cancer (e.g., estrogen receptor positive or negative, progesterone receptor positive or negative, HER-2 positive or negative, triple-negative breast cancer, or BRCA1 and/or BRCA2 positive or negative), lung cancer (e.g., non-small cell lung cancer and small cell lung cancer), endometrial cancer, glioblastoma, B-cell malignancies, biliary tract cancer, bone cancer, choriocarcinoma, connective tissue cancer, cancers of the digestive system, gallbladder cancer, hepatocellular carcinoma, intra-epithelial neoplasm, kidney cancer, liver cancer, lymphoma, skin cancer (e.g., melanoma and basal cell carcinoma), neuroblastoma, mesothelioma, neuroglioma, oral cavity cancer, pediatric cancer, pancreatic endocrine tumors, pituitary adenoma, thymoma, renal cell carcinoma, salivary gland cancer, sarcoma (e.g., Ewing’s sarcoma, fibrosarcoma, and rhabdomyosarcoma), small bowel cancer, ureteral cancer, cancers of the urinary system, and hematological cancers (e.g., acute myeloid leukemia and multiple myeloma).


Exemplary solid cancerous tumors that can be treated with the active agents of the present disclosure include, but are not limited to, a tumor of an organ selected from the group consisting of pancreas, colon, cecum, stomach, brain, liver, gallbladder, head, neck, ovary, kidney, larynx, sarcoma, lung, bladder, uterus, melanoma, prostate, and breast. Exemplary hematological tumors include tumors of the bone marrow, T or B cell malignancies, leukemias, lymphomas, blastomas, myelomas, and the like. Further examples of cancers that may be treated using the methods provided herein include, but are not limited to, lung cancer (including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung), cancer of the peritoneum, gastric or stomach cancer (including gastrointestinal cancer and gastrointestinal stromal cancer), pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, various types of head and neck cancer, and melanoma.


The cancer may specifically be of the following histological type, though it is not limited to these: neoplasm, malignant; carcinoma; carcinoma, undifferentiated; giant and spindle cell carcinoma; small cell carcinoma; papillary carcinoma; squamous cell carcinoma; lymphoepithelial carcinoma; basal cell carcinoma; pilomatrix carcinoma; transitional cell carcinoma; papillary transitional cell carcinoma; adenocarcinoma; gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma; adenoid cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma, familial polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchioloalveolar adenocarcinoma; papillary adenocarcinoma; chromophobe carcinoma; acidophil carcinoma; oxyphilic adenocarcinoma; basophil carcinoma; clear cell adenocarcinoma; granular cell carcinoma; follicular adenocarcinoma; papillary and follicular adenocarcinoma; nonencapsulating sclerosing carcinoma; adrenal cortical carcinoma; endometroid carcinoma; skin appendage carcinoma; apocrine adenocarcinoma; sebaceous adenocarcinoma; ceruminous adenocarcinoma; mucoepidermoid carcinoma; cystadenocarcinoma; papillary cystadenocarcinoma; papillary serous cystadenocarcinoma; mucinous cystadenocarcinoma; mucinous adenocarcinoma; signet ring cell carcinoma; infiltrating duct carcinoma; medullary carcinoma; lobular carcinoma; inflammatory carcinoma; paget’s disease, mammary; acinar cell carcinoma; adenosquamous carcinoma; adenocarcinoma w/squamous metaplasia; thymoma, malignant; ovarian stromal tumor, malignant; thecoma, malignant; granulosa cell tumor, malignant; androblastoma, malignant; sertoli cell carcinoma; leydig cell tumor, malignant; lipid cell tumor, malignant; paraganglioma, malignant; extra-mammary paraganglioma, malignant; pheochromocytoma; glomangiosarcoma; malignant melanoma; amelanotic melanoma; superficial spreading melanoma; lentigo malignant melanoma; acral lentiginous melanomas; nodular melanomas; malignant melanoma in giant pigmented nevus; epithelioid cell melanoma; blue nevus, malignant; sarcoma; fibrosarcoma; fibrous histiocytoma, malignant; myxosarcoma; liposarcoma; leiomyosarcoma; rhabdomyosarcoma; embryonal rhabdomyosarcoma; alveolar rhabdomyosarcoma; stromal sarcoma; mixed tumor, malignant; mullerian mixed tumor; nephroblastoma; hepatoblastoma; carcinosarcoma; mesenchymoma, malignant; brenner tumor, malignant; phyllodes tumor, malignant; synovial sarcoma; mesothelioma, malignant; dysgerminoma; embryonal carcinoma; teratoma, malignant; struma ovarii, malignant; choriocarcinoma; mesonephroma, malignant; hemangiosarcoma; hemangioendothelioma, malignant; Kaposi’s sarcoma; hemangiopericytoma, malignant; lymphangiosarcoma; osteosarcoma; juxtacortical osteosarcoma; chondrosarcoma; chondroblastoma, malignant; mesenchymal chondrosarcoma; giant cell tumor of bone; Ewing’s sarcoma; odontogenic tumor, malignant; ameloblastic odontosarcoma; ameloblastoma, malignant; ameloblastic fibrosarcoma; pinealoma, malignant; chordoma; glioma, malignant; ependymoma; astrocytoma; protoplasmic astrocytoma; fibrillary astrocytoma; astroblastoma; glioblastoma; oligodendroglioma; oligodendroblastoma; primitive neuroectodermal; cerebellar sarcoma; ganglioneuroblastoma; neuroblastoma; retinoblastoma; olfactory neurogenic tumor; meningioma, malignant; neurofibrosarcoma; neurilemmoma, malignant; granular cell tumor, malignant; malignant lymphoma; hodgkin’s disease; hodgkin’s; paragranuloma; malignant lymphoma, small lymphocytic; malignant lymphoma, large cell, diffuse; malignant lymphoma, follicular; mycosis fungoides; other specified non-hodgkin’s lymphomas; B-cell lymphoma; low grade/follicular non-Hodgkin’s lymphoma (NHL); small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; Waldenstrom’s macroglobulinemia; malignant histiocytosis; multiple myeloma; mast cell sarcoma; immunoproliferative small intestinal disease; leukemia; lymphoid leukemia; plasma cell leukemia; erythroleukemia; lymphosarcoma cell leukemia; myeloid leukemia; basophilic leukemia; eosinophilic leukemia; monocytic leukemia; mast cell leukemia; megakaryoblastic leukemia; myeloid sarcoma; hairy cell leukemia; chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); acute myeloid leukemia (AML); and chronic myeloblastic leukemia.


The compounds of the present disclosure may be combined with one or more pharmaceutically-acceptable excipients, including carriers, vehicles, diluents, and adjuvants which may improve solubility, deliverability, dispersion, stability, and/or conformational integrity of the compounds or conjugates thereof.


“Pharmaceutically acceptable salts” means salts of active agent compounds disclosed herein which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include (but are not limited to) acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, 2-naphthalenesulfonic acid, 3-phenylpropionic acid, 4,4′-methylenebis(3-hydroxy-2-ene-1-carboxylic acid), 4-methylbicyclo[2.2.2]oct-2-ene-l-caiboxylic acid, acetic acid, aliphatic mono- and dicarboxylic acids, aliphatic sulfuric acids, aromatic sulfuric acids, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, carbonic acid, cinnamic acid, citric acid, cyclopentanepropionic acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, heptanoic acid, hexanoic acid, hydroxynaphthoic acid, lactic acid, lauryl sulfuric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, muconic acid, o-(4-hydroxybenzoyl)benzoic acid, oxalic acid, p-chlorobenzenesulfonic acid, phenyl-substituted alkanoic acids, propionic acid, p-toluenesulfonic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, tartaric acid, tertiaryhutylacetic acid, trimethylacetic acid, and the like. Pharmaceutically acceptable salts also include (but are not limited to) base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include (but are not limited to) sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include (but are not limited to) ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine and the like. It should be recognized that the particular anion or cation forming a part of any salt of the present disclosure is not critical, so long as the salt, as a whole, is pharmacologically acceptable. Additional non-limiting examples of pharmaceutically acceptable salts and their methods of preparation and use are shown in Handbook of Pharmaceutical Salts: Properties, and Use (P. H. Stahl & C. G. Wermuth eds., Verlag Helvetica Chimica Acta, 2002).


The term “coadministration” refers to administration of two or more active agents, e.g., a cardiac-targeted composition as described herein and another active agent. The timing of coadministration depends in part on the combination and compositions administered and can include administration at the same time, just prior to, or just after the administration of one or more additional therapies. “Coadministration” is meant to include simultaneous or sequential administration of the compound and/or composition individually or in combination. Thus, the preparations can also be combined, when desired, with other active substances (e.g., to reduce metabolic degradation). For example, the compositions described herein can be used in combination with one another, or with other active agents known to be useful in treating MI, and co-occurring conditions thereof.


The active agents of the present disclosure may be present in the pharmaceutical compositions (alone or in combination) at any concentration that allows the pharmaceutical composition to function in accordance with the present disclosure; for example, but not by way of limitation, the compound(s) may be present in a carrier, diluent, or buffer solution in a wt/wt or vol/vol range having a lower level selected from 0.00001%, 0.0001%, 0.005%, 0.001%, 0.005%, 0.01%, 0.05%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1.0%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9% and 2.0%; and an upper level selected from 3%, 3.5%, 4%, 4.5%, 5%, 5.5%, 6%, 6.5%, 7%, 7.5%, 8%, 8.5%, 9%, 9.5%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, and 95%. Non-limiting examples of particular wt/wt or vol/vol ranges include a range of from about 0.0001% to about 95%, a range of from about 0.001% to about 75%; a range of from about 0.005% to about 50%; a range of from about 0.01% to about 40%; a range of from about 0.05% to about 35%; a range of from about 0.1% to about 30%; a range of from about 0.1% to about 25%; a range of from about 0.1% to about 20%; a range of from about 1% to about 15%; a range of from about 2% to about 12%; a range of from about 5% to about 10%; and the like. Any other range that includes a lower level selected from the above-listed lower level concentrations and an upper level selected from the above-listed upper level concentrations also falls within the scope of the present disclosure. Percentages used herein may be weight percentages (wt%) or volume percentages (vol%).


In certain non-limiting embodiments, an effective amount or therapeutic dosage of a pharmaceutical composition of the present disclosure contains, sufficient active agent to deliver from about 0.001 µg/kg to about 100 mg/kg (weight of active agent/body weight of the subject). For example, the composition will deliver about 0.01 µg/kg to about 50 mg/kg, and more particularly about 0.1 µg/kg to about 10 mg/kg, and more particularly about 1 µg/kg to about 1 mg/kg. Practice of a method of the present disclosure may comprise administering to a subject an effective amount of the active agent in any suitable systemic and/or local formulation, in an amount effective to deliver the therapeutic dosage of the active agent. In certain embodiments, an effective dosage may be, in a range of about 1 µg/kg to about 1 mg/kg of the active agent.


In certain non-limiting embodiments, an effective amount or therapeutic dosage of a pharmaceutical composition of the present disclosure contains a heteroarotinoid (e.g., SHetA2) in a dose range that will result in 0.01 micromolar to 100 millimolar blood or tissue levels, and a Azabicyclooctan-3-one derivative, and/or CDK4/6 inhibitor in a concentration in a dose range that will result in 0.01 micromolar to 100 millimolar blood or tissue levels. In certain non-limiting embodiments, an effective amount or therapeutic dosage of a pharmaceutical composition of the present disclosure contains SHetA2 in a concentration in a dose range that will result in 0.01 micromolar to 100 millimolar, and a second active agent (e.g., Palbociclib) in a dose range that will result in 0.01 micromolar to 100 millimolar blood or tissue levels.


In non-limiting embodiments, the synergistic ratio of the compositions of the first active agent (heteroarotinoid) and the second active agent (e.g., see drug combinations in Tables 10-13) may be in a range from 50:1 wt/wt to 1:50 wt/wt, 45:1 wt/wt to 1:45 wt/wt, 40:1 wt/wt to 1:40 wt/wt, 35:1 wt/wt to 1:35 wt/wt, 30:1 wt/wt to 1:30 wt/wt, 25:1wt/wt to 1:25 wt/wt, e.g., 25:1, 24:1, 23:1, 22:1, 21:1, 20:1, 19:1, 18:1, 17:1, 16:1, 15:1, 14:1, 13:1, 12:1, 11:1, 10:1,9:1,8:1,7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1:21, 1:22, 1:23, 1:24, or 1:25 or may be in any range bounded by the ratios listed above.


The term “synergistic” or “synergistic effect” or “synergistic interaction” as used herein refers to a therapeutic combination which is more effective than the additive effects of the two or more single active agents. A “synergistic ratio” is a ratio of two compounds which results in a synergistic effect. A determination of a synergistic interaction between the active agents described herein may be based on the results obtained from the assays described herein. The results of these assays can be analyzed using the Chou and Talalay combination method (Chou TC, Talalay P. “Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors.” Adv Enzyme Regul. 1984;22: 27-55) and Dose-Effect Analysis with CalcuSyn software in order to obtain a Combination Index. The combinations provided herein have been evaluated in several assay systems, and the data can be analyzed utilizing a standard program for quantifying synergism, additivism, and antagonism among anticanceragents. An example program is that described by Chou and Talalay, in “New Avenues in Developmental Cancer Chemotherapy,” Academic Press, 1987, Chapter 2. Combination Index values less than 0.9 indicate synergy, values greater than 1.2 indicate antagonism and values between 0.9 to 1.1 indicate additive effects (e.g., see Table 4 below). The combination therapy may provide “synergy” and prove “synergistic”, i.e., the effect achieved when the active agents used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active agents are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered in succession (“alternation therapy”) or in parallel as separate formulations; or (3) by some other effective regimen. When delivered in successive administrations, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.


Certain non-limiting embodiments of the present disclosure are directed to a method that comprises administering to a subject in need thereof any of the pharmaceutical compositions disclosed or otherwise contemplated herein.


The pharmaceutical compositions may be administered via any mechanisms disclosed herein or otherwise contemplatable by a person having ordinary skill in the art. In one non-limiting embodiment, the administration occurs via an inhaler, which aerosolizes the active agents.


The pharmaceutical compositions of the present disclosure may be administered for any purpose disclosed or otherwise contemplated herein, as well as for any purpose within the purview of a person having ordinary skill in the art. In one non-limiting embodiment, the pharmaceutical compositions are administered in a method of treating or reducing the occurrence of cancer. However, this treatment method is not to be construed as limiting of the present disclosure, and any diseases, disorders, or conditions disclosed herein or otherwise contemplatable by a person having ordinary skill in the art (given the subject application) which may derive a therapeutic effect by treatment with the compositions disclosed herein also fall within the scope of the methods of the present disclosure.


Practice of the methods of the present disclosure may comprise administering to a subject therapeutically effective amounts of the active agents in any suitable systemic and/or local formulation, in an amount effective to deliver the dosages listed herein. The dosage can be administered, for example but not by way of limitation, on a one-time basis, or administered at multiple times (for example but not by way of limitation, from one to five times per day, or once or twice per week), or continuously via a venous drip, depending on the desired therapeutic effect. In one non-limiting example of a therapeutic method of the present disclosure, the active agent is provided in an IV infusion in the range of from about 0.01 mg/kg to about 10 mg/kg of body weight once a day.


The compositions and dosage forms of the present disclosure can be administered in a single dose treatment or in multiple dose treatments on a schedule and over a time period appropriate to the age, weight, and condition of the subject, the particular composition used, and the route of administration. In one non-limiting embodiment, a single dose of the composition according to the disclosure is administered. In other non-limiting embodiments, multiple doses are administered. The frequency of administration can vary depending on any of a variety of factors, e.g., severity of the symptoms, or whether the composition is used for prophylactic or curative purposes. For example, in certain non-limiting embodiments, the composition is administered once per month, twice per month, three times per month, every other week, once per week, twice per week, three times per week, four times per week, five times per week, six times per week, every other day, daily, twice a day, or three times a day. The duration of treatment (i.e., the period of time over which the composition is administered) can vary, depending on any of a variety of factors, e.g., subject response. For example, the composition can be administered over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more. Where used herein, unless otherwise indicated, the dosage amount refers to the amount of active pharmaceutical ingredient (API) that is administered to the subject.


The dosage of an administered active agent for humans will vary depending upon factors such as (but not limited to) the patient’s age, weight, height, sex, general medical condition, and previous medical history. In certain non-limiting embodiments, where the active agent is administered by injection or infusion, the recipient may be provided with a dosage of the active agent that is in the range of from about 1 mg to about 1000 mg, and it may be administerted as a single infusion or multiple injections, although a lower or higher dosage also may be administered. In certain non-limiting embodiments, the dosage may be in the range of from about 25 mg to about 100 mg of the active agent per square meter (m2) of body surface area for a typical adult, although a lower or higher dosage also may be administered. Non-limiting examples of dosages of the active agent that may be administered to a human subject include, but are not limited to, those in ranges of 1 to 1000 mg, 1 to 600 mg, 1 to 500 mg, 1 to 400 mg, 1 to 300 mg, 1 to 200 mg, 100 to 600 mg, 100 to 500 mg, 100 to 400 mg, 100 to 300 mg, 100 to 200 mg, 150 to 600 mg, 150 to 500 mg, 150 to 400 mg, 150 to 300 mg, 150 to 250 mg, 150 to 200 mg, 200 to 7500 mg, 200 to 600 mg, 200 to 500 mg, 200 to 400 mg, 200 to 300 mg, and 200 to 250 mg, or any subrange within any of the aforementioned ranges. Dosages may be repeated as needed, for example (but not by way of limitation), once per week for 4-10 weeks, once per week for 8 weeks, or once per week for 4 weeks. It may also be given less frequently, such as (but not limited to) every other week for several months, or more frequently, such as twice weekly or by continuous infusion.


In certain non-limiting embodiments, the present disclosure is directed to a dosing regimen comprising multiple dosing cycles (e.g., wherein the first dosing cycle is a step-up, fractionated dosing cycle). In some non-limiting embodiments, the dose may range from 50 mg to 200 mg (e.g., from 50 mg to 175 mg, from 50 mg to 150 mg, from 50 mg to 125 mg, from 50 mg to 100 mg, from 50 mg to 75 mg, from 50 mg to 70 mg, from 52 mg to 100 mg, from 52 mg to 75 mg, from 50 mg to 180 mg, from 55 mg to 150 mg, from 55 mg to 100 mg, from 55 mg to 70 mg, from 55 mg to 65 mg, from 58 mg to 62 mg; e.g., about 60 mg,,or any subrange within any of the aforementioned ranges). In some non-limiting embodiments, the dose may be about 60 mg. In some non-limiting embodiments, the dose is about 1 mg. In some non-limiting embodiments, the dose is about 2 mg.


In some non-limiting embodiments, the dose is from 20 mg to 200 mg (e.g., from 20 mg to 175 mg, from 20 mg to 150 mg, from 20 mg to 100 mg, from 20 mg to 75 mg, from 30 mg to 175 mg, from 40 mg to 175 mg, from 45 mg to 175 mg, from 50 mg to 175 mg, from 30 mg to 150 mg, from 40 mg to 100 mg, from 45 mg to 75 mg, from 50 mg to 70 mg, from 55 mg to 65 mg, from 58 mg to 62 mg; about 20 mg, about 30 mg, about 45 mg, or e.g., about 60 mg, or any subrange within any of the aforementioned ranges). In some non-limiting embodiments, the dose is from about 12 mg to about 48 mg (e.g., from about 12 mg to about 42 mg, from about 12 mg to about 36 mg, from about 12 mg to about 30 mg, from about 18 mg to about 48 mg, from about 18 mg to about 42 mg, from about 24 mg to about 42 mg, from about 27 mg to about 42 mg, from about 24 mg to about 36 mg, from about 27 mg to about 33 mg, from about 28 mg to about 32 mg; e.g., about 24 mg, about 27 mg, about 30 mg, about 33 mg, or about 36 mg, or any subrange within any of the aforementioned ranges).


In some non-limiting embodiments, the dosing regimen comprises administration of a dose in a range of from 100 mg to 750 mg (e.g., from 100 mg to 725 mg, from 100 mg to 700 mg, from 100 mg to 675 mg, from 100 mg to 650 mg, from 100 mg to 625 mg, from 100 mg to 600 mg, from 100 mg to 575 mg, from 100 mg to 550 mg, from 100 mg to 525 mg, from 100 mg to 500 mg, from 100 mg to 475 mg, from 100 mg to 450 mg, from 100 mg to 425 mg, from 100 mg to 400 mg, from 100 mg to 375 mg, from 100 mg to 350 mg, from 100 mg to 325 mg, from 100 mg to 300 mg, from 100 mg to 275 mg, from 100 mg to 250 mg, or from 100 mg to 225 mg, from 100 mg to 200 mg, from 100 mg to 175 mg, from 100 mg to 150 mg, or from 100 mg to 125 mg, or any subrange within any of the aforementioned ranges).


In some non-limiting embodiments, the dosing regimen comprises administration of a dose in a range of from 200 mg to 750 mg (e.g., from 200 mg to 725 mg, from 200 mg to 700 mg, from 200 mg to 675 mg, from 200 mg to 650 mg, from 200 mg to 625 mg, from 200 mg to 600 mg, from 200 mg to 575 mg, from 200 mg to 550 mg, from 200 mg to 525 mg, from 200 mg to 500 mg, from 200 mg to 475 mg, from 200 mg to 450 mg, from 200 mg to 425 mg, from 200 mg to 400 mg, from 200 mg to 375 mg, from 200 mg to 350 mg, from 200 mg to 325 mg, from 200 mg to 300 mg, from 200 mg to 275 mg, from 200 mg to 250 mg, or from 200 mg to 225 mg, or any subrange within any of the aforementioned ranges).


In some non-limiting embodiments, the dosing regimen comprises administration of a dose in a range of from 300 mg to 750 mg (e.g., from 300 mg to 725 mg, from 300 mg to 700 mg, from 300 mg to 675 mg, from 300 mg to 650 mg, from 300 mg to 625 mg, from 300 mg to 600 mg, from 300 mg to 575 mg, from 300 mg to 550 mg, from 300 mg to 525 mg, from 300 mg to 500 mg, from 300 mg to 475 mg, from 300 mg to 450 mg, from 300 mg to 425 mg, from 300 mg to 400 mg, from 300 mg to 375 mg, from 300 mg to 350 mg, or from 300 mg to 325 mg, or any subrange within any of the aforementioned ranges).


In some non-limiting embodiments, the dosing regimen comprises administration of a dose in a range of from 400 mg to 750 mg (e.g., from 400 mg to 725 mg, from 400 mg to 700 mg, from 400 mg to 675 mg, from 400 mg to 650 mg, from 400 mg to 625 mg, from 400 mg to 600 mg, from 400 mg to 575 mg, from 400 mg to 550 mg, from 400 mg to 525 mg, from 400 mg to 500 mg, from 400 mg to 475 mg, from 400 mg to 450 mg, or from 400 mg to 425 mg, or any subrange within any of the aforementioned ranges).


In some non-limiting embodiments, the dosing regimen comprises administration of a dose in a range of from 500 mg to 750 mg (e.g., from 500 mg to 725 mg, from 500 mg to 700 mg, from 500 mg to 675 mg, from 500 mg to 650 mg, from 500 mg to 625 mg, from 500 mg to 600 mg, from 500 mg to 575 mg, from 500 mg to 550 mg, or from 500 mg to 525 mg, or any subrange within any of the aforementioned ranges).


In some non-limiting embodiments, the dosing regimen comprises administration of a dose in a range of from 600 mg to 750 mg (e.g., from 600 mg to 725 mg, from 600 mg to 700 mg, from 600 mg to 675 mg, from 600 mg to 650 mg, from 600 mg to 625 mg, or any subrange within any of the aforementioned ranges).


In some non-limiting embodiments, the active agent is provided in a concentration of about 1 nM, about 5 nM, about 10 nM, about 25 nM, about 50 nM, about 75 nM, about 100 nM, about 150 nM, about 200 nM, about 250 nM, about 300 nM, about 350 nM, about 400 nM, about 500 nM, about 550 nM, about 600 nM, about 700 nM, about 800 nM, about 900 nM, about 1 µM, about 2 µM, about 3 µM, about 4 µM,about 5 µM, about 6 µM,about 7 µM, about 8 µM,about 9 µM, about 10 µM, about 15 µM,about 20 µM, about 25 µM, about 30 µM, about 35 µM, about 40 µM, about 45 µM, about 50 µM, about 60 µM, about 70 µM, about 75 µM, about 80 µM, about 90 µM, about 100 µM, about 125 µM, about 150 µM, about 175 µM, about 200 µM, about 250 µM, about 300 µM, about 350 µM, about 400 µM, about 500 µM, about 600 µM, about 700 µM, about 750 µM, about 800 µM, about 900 µM, about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 11 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 55 mM, about 60 mM, about 65 mM, about 70 mM, about 75 mM, about 80 mM, about 85 mM, about 90 mM, about 95 mM, about 100 mM, about 100 mM, about 110 mM, about 120 mM, about 130 mM, about 140 mM, about 150 mM, about 160 mM, about 170 mM, about 180 mM, about 190 mM, about 200 mM, about 250 mM, about 300 mM, about 400 mM, about 500 mM, about 600 mM, about 700 mM, about 800 mM, about 900 mM, about 1000 mM, about 1 M, about 1.1 M, about 1.2 M, about 1.3 M, about 1.4 M, about 1.5 M, about 1.6 M, about 1.7 M, about 1.8 M, about 1.9 M, about 2 M, about 3 M, about 4 M, about 5 M, about 6 M, about 7 M, about 8 M, about 9 M, about 10 M, about 15 M, about 20 M, about 25 M, about 30 M, about 35 M, about 40 M, about 45 M, about 50 M, about 75 M, about 100 M, or any range in between any two of the aforementioned concentrations, including said two concentrations as endpoints of the range, or any number in between any two of the aforementioned concentrations.


When administered orally, the active agent composition may be protected from digestion. This can be accomplished either by complexing the active agent with a composition to render it resistant to acidic and enzymatic hydrolysis or by packaging the active agent in an appropriately resistant carrier such as (but not limited to) a liposome, e.g., such as shown in U.S. Pat. No. 5,391,377.


In certain embodiments, the different therapeutic compounds of the disclosure can be administered within one hour of each other, within two hours of each other, within 3 hours of each other, within 6 hours of each other, within 12 hours of each other, within 24 hours of each other, within 36 hours of each other, within 48 hours of each other, within 72 hours of each other, or more. Thus an individual who receives such treatment can benefit from a combined effect of the different therapeutic compounds.


The active agents of the present disclosure can be administered to a subject by any of a number of effective routes of administration including, for example, orally (for example, drenches as in aqueous or non-aqueous solutions or suspensions, tablets, capsules (including sprinkle capsules and gelatin capsules), boluses, powders, granules, pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingually); anally, rectally or vaginally (for example, as a pessary, cream or foam); parenterally (including intramuscularly, intravenously, subcutaneously or intrathecally as, for example, a sterile solution or suspension); nasally; intraperitoneally; subcutaneously; transdermally (for example as a patch applied to the skin); and topically (for example, as a cream, ointment or spray applied to the skin, or as an eye drop). The compounds may also be formulated for inhalation. In certain embodiments, a compound may be simply dissolved or suspended in sterile water. Oral formulations may be formulated such that the active agents passes through a portion of the digestive system before being released, for example it may not be released until reaching the small intestine, or the colon. Details of appropriate routes of administration and compositions suitable for same can be found in, for example, U.S. Pat. Nos. 6,110,973, 5,763,493, 5,731,000, 5,541,231, 5,427,798, 5,358,970 and 4,172,896, as well as in patents cited therein.


Tablets, and other solid dosage forms of the pharmaceutical compositions, such as dragees, capsules (including sprinkle capsules and gelatin capsules), pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical- formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.


Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.


Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.


Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.


Formulations of the pharmaceutical compositions for rectal, vaginal, or urethral administration may be presented as a suppository, which may be prepared by mixing one or more active compounds with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.


Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. In certain embodiments, the active agents of the present disclosure can be formulated into suppositories, slow release formulations, or intrauterine delivery devices (IUDs).


Formulations of the pharmaceutical compositions for administration to the mouth may be presented as a mouthwash, or an oral spray, or an oral ointment.


Alternatively or additionally, compositions can be formulated for delivery via a catheter, stent, wire, or other intraluminal device. Delivery via such devices may be especially useful for delivery to the bladder, urethra, ureter, rectum, or intestine.


Dosage forms for the topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required. The ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the active compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.


Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. Exemplary ophthalmic formulations are described in U.S. Publication Nos. 2005/0080056, 2005/0059744, 2005/0031697 and 2005/004074 and U.S. Pat. No. 6,583,124, the contents of which are incorporated herein by reference. If desired, liquid ophthalmic formulations have properties similar to that of lacrimal fluids, aqueous humor or vitreous humor or are compatable with such fluids. A preferred route of administration is local administration (e.g., topical administration, such as eye drops, or administration via an implant).


The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.


For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated can be used in the formulation. Such penetrants are generally known in the art, and include, e.g., for transmucosal administration, bile salts and fusidic acid derivatives. In addition, detergents can be used to facilitate permeation. Transmucosal administration can be through nasal sprays or using suppositories. For topical transdermal administration, the agents are formulated into ointments, creams, salves, powders, and gels. Transdermal delivery systems can also include (for example but not by way of limitation) patches. The present compositions can also be administered in sustained delivery or sustained release mechanisms. For example, biodegradeable microspheres or capsules or other biodegradeable polymer configurations capable of sustained delivery can be included herein.


The compositions of the present disclosure may be formulated as implants, in the form of either biodegradable microparticles or small squared films comprising the microparticles of the present disclosure. The following describes methods of making such implants. Microparticles (e.g., 5-100 micrometers) containing different loadings of the compounds of the present disclosure, such as heteroarotinoids, are prepared by spray drying suspensions of the nanocrystals of the compounds and a biodegradable polymer (for example, polylactic acid of molecular weights 50,000 -100,000) or polylactic-co-glycolic acid copolymer (e.g., proportions 75:25 or 50:50). Microparticles may contain, e.g., 10 - 50% wt/wt drug:polymer and can be implanted alone, or in a biodegradable film, e.g., as an implantable chitosan-egg phosphatidylcholine (ePC) films. To make such chitosan-egg phosphatidylcholine (ePC) films, chitosan flakes and ePC can be dissolved in a 1% acetic acid at a ratio of 1:0.8 (wt/wt). Microparticles containing the drug in nanocrystal form are dispersed in the chitosan-ePC solution in different proportions (e.g., 1:3, 1:5, 1:7 and 1:10 wt/wt) to achieve the release of different drug doses. The resulting microparticle-chitosan-ePC suspension can be poured into a Teflon dish to have a 2-3 mm thickness and allowed to dry in a covered dessicator for 5 days. After the films are dry, they can be cut into small squares of 15×15 mm2. The implants can be made in different forms, including but not limited to thin films, rods, and wafers. Other biodegradable polymers that can be used to make implants include, but are not limited to, poly-lactic acid, poly-lactic-co-glycolic acid copolymer, poly-caprolactone, poly-sebacic acid, poly-adipic acid, poly (3-hydroxybutyric acid), poly(3-hydroxybutyrate-co-3-hydroxyvalerate, poly-trimethylene carbonate, chitosan, chitin, gelatin, collagen, and hyaluronic acid.


In non-limiting embodiments, gels comprising the active agents of the present disclosure can be made by combining the active agents in various proportions to a sodium alginate gel base or carbomer jelly base to form a homogeneous gel suitable for topical application.


In non-limiting embodiments, ointments comprising the active agents of the present disclosure can be made by combining the active agents in various proportions to a Hydrophilic Petrolatum USP base, Lanolin, USP base or to Polyethylene glycol ointment, NF to form a homogeneous ointment suitable for topical application.


In non-limiting embodiments, creams comprising the active agents of the present disclosure can be made by combining the active agents in various proportions in suspension in water and glycerin (e.g., 20:1 parts) and emulsified in a mixture of e.g., Lanolin, Beeswax USP-NF and Cetyl alcohol, plus a Tween 80 and Span 80.


Several gel, ointment, and/or cream compositions that can be used are shown in García-Contreras L, Abu-Izza K, Lu DR. “Biodegradable cisplatin microspheres for direct brain injection: Preparation and characterization.” Pharm Dev Technol (1997) 2(1): 53-65.


For inhalation, the present compositions can be delivered using any system known in the art, including (but not limited to) dry powder aerosols, liquids delivery systems, air jet nebulizers, propellant systems, and the like. For example (but not by way of limitation), the pharmaceutical formulation can be administered in the form of an aerosol or mist. For aerosol administration, the formulation can be supplied in finely divided form along with a surfactant and propellant. In another non-limiting aspect, the device for delivering the formulation to respiratory tissue is an inhaler in which the formulation vaporizes. Other liquid delivery systems include (for example but not by way of limitation) air jet nebulizers.


For inhalation, the present compositions can be delivered using any system known in the art, including (but not limited to) dry powder aerosols, liquids delivery systems, air jet nebulizers, propellant systems, and the like. For example (but not by way of limitation), the pharmaceutical formulation can be administered in the form of an aerosol or mist. For aerosol administration, the formulation can be supplied in finely divided form along with a surfactant and propellant. In another non-limiting aspect, the device for delivering the formulation to respiratory tissue is an inhaler in which the formulation vaporizes. Other liquid delivery systems include (for example, but not by way of limitation) air jet nebulizers.


Pharmaceutical compositions suitable for parenteral administration comprise one or more active compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.


Examples of suitable aqueous and nonaqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.


These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.


In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.


Injectable depot forms are made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue.


As noted, effective amounts of the active agents may be administered orally, in the form of a solid or liquid preparations such as capsules, pills, tablets, lozenges, melts, powders, suspensions, solutions, elixirs or emulsions. Solid unit dosage forms can be capsules of the ordinary gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, and cornstarch, or the dosage forms can be sustained release preparations. The pharmaceutical composition may contain a solid carrier, such as a gelatin or an adjuvant. The tablet, capsule, and powder may contain from about 0.05 to about 95% of the active substance compound by dry weight. When administered in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils may be added. The liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol. When administered in liquid form, the pharmaceutical composition particularly contains from about 0.005 to about 95% by weight of the active agent(s). For example, a dose of about 10 mg to about 1000 mg once or twice a day could be administered orally.


In another embodiment, the active agents of the present disclosure can be tableted with conventional tablet bases such as lactose, sucrose, and cornstarch in combination with binders, such as acacia, cornstarch, or gelatin, disintegrating agents such as potato starch or alginic acid, and a lubricant such as stearic acid or magnesium stearate. Liquid preparations are prepared by dissolving the active agents in an aqueous or non-aqueous pharmaceutically acceptable solvent which may also contain suspending agents, sweetening agents, flavoring agents, and preservative agents as are known in the art.


In one non-limiting aspect, the active agent is incorporated in lipid monolayers or bilayers, such as (but not limited to) liposomes. Liposomes and liposomal formulations can be prepared according to standard methods and are also well known in the art, such as (but not limited to) those disclosed in U.S. Pat. Nos. 6,110,490; 6,096,716; 5,283,185; 5,279,833; 4,235,871; 4,501,728; and 4,837,028.


In one non-limiting aspect, the compositions are prepared with carriers that will protect the active agent against rapid elimination from the body, such as (but not limited to) a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as (but not limited to) ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.


The active agents in general may be formulated to obtain compositions that include one or more pharmaceutically suitable excipients, surfactants, polyols, buffers, salts, amino acids, or additional ingredients, or some combination of these. This can be accomplished by known methods to prepare pharmaceutically useful dosages, whereby the active agent is combined in a mixture with one or more pharmaceutically suitable excipients. Sterile phosphate-buffered saline is one non-limiting example of a pharmaceutically suitable excipient.


The acid addition salts may include, but are not limited to, 4-acetamidobenzoate, acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate (besylate), benzoate, bisulfate, bitartrate, butyrate, calcium edetate, camphorate, cam phorsulfonate (camsylate), caprate (decanoate), caproate (hexanoate), caprylate (octanoate), cinnamate, citrate, cyclamate, digluconate, 2,5-dihydroxybenzoate, disuccinate, dodecylsulfate (estolate), edetate (ethylenediaminetetraacetate), estolate (lauryl sulfate), ethane-1,2-disulfonate (edisylate), ethanesulfonate (esylate), formate, fumarate, galactarate (mucate), gentisate (2,5-dihydroxybenzoate), glucoheptonate (gluceptate), gluconate, glucuronate, glutamate, glutarate, glycerophosphorate, glycolate, hexylresorcinate, hippurate, hydrabamine (N,N?-di(dehydroabietyl)-ethylenediamine), hydrobromide, hydrochloride, hydroiodide, hydroxynaphthoate, isobutyrate, lactate, lactobionate, laurate, malate, maleate, malonate, mandelate, methanesulfonate (mesylate), methylsulfate, mucate, naphthalene-1,5-disulfonate (napadisylate), naphthalene-2-sulfonate (napsylate), nicotinate, nitrate, oleate, palmitate, p-aminobenzenesulfonate, p-aminosalicyclate, pamoate (embonate), pantothenate, pectinate, persulfate, phenylacetate, phenylethylbarbiturate, phosphate, polygalacturonate, propionate, p-toluenesulfonate (tosylate), pyroglutamate, pyruvate, salicylate, sebacate, stearate, subacetate, succinate, sulfamate, sulfate, tannate, tartrate, teoclate (8-chlorotheophyllinate), thiocyanate, triethiodide, undecanoate, undecylenate, and valerate.


Non-limiting examples of routes of administration of the compositions described herein include parenteral injection, e.g., by subcutaneous, intramuscular, or transdermal delivery. Other forms of injection include (but are not limited to) intravenous, intraarterial, intralymphatic, intrathecal, intraocular, intranasal, intracranial, intracerebral, intraperitoneal, or intracavitary injection. In parenteral administration, the compositions will be formulated in a unit dosage injectable form such as (but not limited to) a solution, suspension, or emulsion, in association with a pharmaceutically acceptable excipient. Such excipients are inherently nontoxic and nontherapeutic. Non-limiting examples of such excipients include saline, Ringer’s solution, dextrose solution, and Hanks’ solution. Nonaqueous excipients such as (but not limited to) fixed oils and ethyl oleate may also be used. An alternative non-limiting excipient is 5% dextrose in saline. The excipient may contain minor amounts of additives such as (but not limited to) substances that enhance isotonicity and chemical stability, including buffers and preservatives. The active agents can be delivered or administered alone or as pharmaceutical compositions by any means known in the art, such as (but not limited to) systemically, regionally, or locally, for example by intraarterial, intrathecal (IT), intravenous (IV), parenteral, intrapleural cavity, topical, oral, or local administration, as subcutaneous, intratracheal (e.g., by aerosol) or transmucosal administration (e.g., buccal, bladder, vaginal, uterine, rectal, and/or nasal mucosa). Administration can also be localized directly into a tumor. Administration into the systemic circulation by intravenous, inhalation, mucosal, or subcutaneous administration is typical. Intravenous administration can be, for example (but not by way of limitation), by infusion over a period such as (but not limited to) 30-90 min or by a single bolus injection, or by other regimens as described elsewhere herein.


For parenteral administration, for example, the active agents may be dissolved in a physiologically acceptable pharmaceutical carrier and administered as either a solution or a suspension. Illustrative of suitable pharmaceutical carriers are water, saline, dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative, or synthetic origin. The pharmaceutical carrier may also contain preservatives and buffers as are known in the art.


When an effective amount of the active agents is administered by intravenous, cutaneous, or subcutaneous injection, the compound is particularly in the form of a pyrogen-free, parenterally acceptable aqueous solution or suspension. The preparation of such parenterally acceptable solutions, having due regard to pH, isotonicity, stability, and the like, is well within the skill in the art. A particular pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection may contain, in addition to the active agent, an isotonic vehicle such as Sodium Chloride Injection, Ringer’s Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer’s Injection, or other vehicle as known in the art. The pharmaceutical compositions of the present disclosure may also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.


As noted, particular amounts and modes of administration can be determined by one skilled in the art. One skilled in the art of preparing formulations can readily select the proper form and mode of administration, depending upon the particular characteristics of the active agents selected, the condition to be treated, the stage of the condition, and other relevant circumstances using formulation technology known in the art, described, for example, in Remington: The Science and Practice of Pharmacy, 22nd ed.


Additional pharmaceutical methods may be employed to control the duration of action of the active agents. Increased half-life and/or controlled release preparations may be achieved through the use of proteins or polymers to conjugate, complex with, and/or absorb the active agents as discussed previously herein. The controlled delivery and/or increased half-life may be achieved by selecting appropriate macromolecules (for example but not by way of limitation, polysaccharides, polyesters, polyamino acids, homopolymers, polyvinyl pyrrolidone, ethylenevinylacetate, methylcellulose, or carboxymethylcellulose, and acrylamides such as N-(2-hydroxypropyl) methacrylamide), and the appropriate concentration of macromolecules as well as the methods of incorporation, in order to control release.


Another possible method useful in controlling the duration of action of the active agents by controlled release preparations and half-life is incorporation of the active agents or their functional derivatives into particles of a polymeric material such as polyesters, polyamides, polyamino acids, hydrogels, poly(lactic acid), ethylene vinylacetate copolymers, copolymer micelles of, for example, polyethylene glycol (PEG) and poly(1-aspartamide).


Additional pharmaceutical methods may be employed to increase bioavailability of the drug, such as Kolliphor HS15.


It is also possible to entrap the active agents in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (for example, hydroxymethylcellulose or gelatine-microcapsules and poly-(methylmethacylate) microcapsules, respectively), in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles, and nanocapsules), or in macroemulsions. Such techniques are well known to persons having ordinary skill in the art.


When the active agents are to be used as an injectable material, they can be formulated into a conventional injectable carrier. Suitable carriers include biocompatible and pharmaceutically acceptable phosphate buffered saline solutions, which are particularly isotonic.


For reconstitution of a lyophilized product in accordance with the present disclosure, one may employ a sterile diluent, which may contain materials generally recognized for approximating physiological conditions and/or as required by governmental regulation. In this respect, the sterile diluent may contain a buffering agent to obtain a physiologically acceptable pH, such as sodium chloride, saline, phosphate-buffered saline, and/or other substances which are physiologically acceptable and/or safe for use. In general, the material for intravenous injection in humans should conform to regulations established by the Food and Drug Administration, which are available to those in the field. The pharmaceutical composition may also be in the form of an aqueous solution containing many of the same substances as described above for the reconstitution of a lyophilized product.


The active agents can also be administered as a pharmaceutically acceptable acid- or base-addition salt, formed by reaction with inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, tauric acid, maleic acid, and fumaric acid, or by reaction with an inorganic base such as sodium hydroxide, ammonium hydroxide, potassium hydroxide, and organic bases such as monoalkyl, dialkyl, trialkyl and aryl amines, and substituted ethanolamines.


In certain embodiments, the present disclosure includes an active agent composition wherein at least one of the active agents is coupled (e.g., by covalent bond) directly or indirectly to a carrier molecule.


Formulated compositions comprising the active agents of the present disclosure can be provided in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. Compositions can also take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.


In some non-limiting methods, the patient is administered the active agent every one, two, three, or four weeks, for example. The dosage depends on the frequency of administration, condition of the patient, response to prior treatment (if any), whether the treatment is prophylactic or therapeutic, and whether the disorder is acute or chronic, among other factors.


The number of dosages administered may depends on the severity and temporal nature of the disorder (e.g., whether presenting acute or chronic symptoms) and the response of the disorder to the treatment. For acute disorders or acute exacerbations of a chronic disorder, between 1 and 10 doses may be used. Sometimes a single bolus dose, optionally in divided form, is sufficient for an acute disorder or acute exacerbation of a chronic disorder. Treatment can be repeated for recurrence of an acute disorder or acute exacerbation. For chronic disorders, the active agent may be administered at regular intervals, such as (but not limited to) weekly, fortnightly, monthly, quarterly, every six months for at least 1, 5, or 10 years, or for the life of the patient.


Compositions can be administered in a single dose treatment or in multiple dose treatments on a schedule and over a time period appropriate to the age, weight, and condition of the subject, the particular composition used, and the route of administration. In one non-limiting embodiment, a single dose of the composition according to the disclosure is administered. In other non-limiting embodiments, multiple doses are administered. The frequency of administration can vary depending on any of a variety of factors, e.g., severity of the symptoms, degree of immunoprotection desired, or whether the composition is used for prophylactic or curative purposes. For example, in certain non-limiting embodiments, the composition is administered once per month, twice per month, three times per month, every other week, once per week, twice per week, three times per week, four times per week, five times per week, six times per week, every other day, daily, twice a day, or three times a day. The duration of treatment (i.e., the period of time over which the composition is administered) can vary, depending on any of a variety of factors, e.g., subject response. For example (but not by way of limitation), the composition can be administered over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.


The dosage of an administered active agent for humans will vary depending upon factors such as (but not limited to) the patient’s age, weight, height, sex, general medical condition, and previous medical history. A dosage may be provided as several smaller amounts. For example, a single dosage of 500 mg may be administered as ten 50 mg tablets or capsules, or as five 100 mg tablets or capsules. The amounts of doses or dosages described herein may be provided in a single capsule, tablet, injection, infusion, or other more of delivery. Or, the amounts of drug which comprise the doses or dosages described herein may be provided in two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12) capsules, tablets, injections, infusions, or other modes of delivery.


In certain non-limiting embodiments, the recipient may be provided with a dosage of the active agent that is in the range of from about 1 mg to about 1000 mg. A lower or higher dosage also may be administered. In certain non-limiting embodiments, the dosage may be in the range of from about 25 mg to about 100 mg of the active agent per square meter (m2) of body surface area for a typical adult, although a lower or higher dosage also may be administered. Non-limiting examples of dosages of the active agent that may be administered to a human subject further include 1 to 750 mg, 1 to 600 mg, 1 to 500 mg, 1 to 400 mg, 1 to 300 mg, 1 to 200 mg, or 1 to 100 mg, although higher or lower doses may be used. Dosages may be repeated as needed, for example (but not by way of limitation), once per week for 4-10 weeks, once per week for 8 weeks, or once per week for 4 weeks. In certain non-limiting embodiments, the dosage can be provided as an infusion, for example as a single injection or as multiple injections. It may also be given less frequently, such as (but not limited to) every other week for several months, or more frequently, such as twice weekly, or by continuous infusion.


In some non-limiting embodiments, a per capsule or tablet dose may range from 25 mg to 200 mg (e.g., from 25 mg to 175 mg, from 25 mg to 150 mg, from 25 mg to 125 mg, from 25 mg to 100 mg, from 25 mg to 75 mg, from 25 mg to 70 mg, from 40 mg to 100 mg, from 40 mg to 75 mg, from 40 mg to 175 mg, from 40 mg to 150 mg, from 40 mg to 125 mg, from 40 mg to 70 mg, from 40 mg to 60 mg, or from 45 mg to 55 mg). In some non-limiting embodiments, the dose per capsule or tablet may be about 50 mg.


In some non-limiting embodiments, the dose per capsule or tablet is from 20 mg to 200 mg (e.g., from 20 mg to 175 mg, from 20 mg to 150 mg, from 20 mg to 100 mg, from 20 mg to 75 mg, from 30 mg to 175 mg, from 40 mg to 175 mg, from 45 mg to 175 mg, from 50 mg to 175 mg, from 30 mg to 150 mg, from 40 mg to 100 mg, from 45 mg to 75 mg, from 50 mg to 70 mg, from 55 mg to 65 mg, from 58 mg to 62 mg; about 20 mg, about 30 mg, about 45 mg, or e.g., about 60 mg). In some non-limiting embodiments, the dose is from about 12 mg to about 48 mg (e.g., from about 12 mg to about 42 mg, from about 12 mg to about 36 mg, from about 12 mg to about 30 mg, from about 18 mg to about 48 mg, from about 18 mg to about 42 mg, from about 24 mg to about 42 mg, from about 27 mg to about 42 mg, from about 24 mg to about 36 mg, from about 27 mg to about 33 mg, from about 28 mg to about 32 mg; e.g., about 24 mg, about 27 mg, about 30 mg, about 33 mg, or about 36 mg).


In some non-limiting embodiments, the dosing regimen comprises administration of a loading dose, such as from 20 mg to 200 mg (e.g., from 20 mg to 175 mg, from 20 mg to 150 mg, from 20 mg to 100 mg, from 20 mg to 75 mg, from 30 mg to 175 mg, from 40 mg to 175 mg, from 45 mg to 175 mg, from 50 mg to 175 mg, from 30 mg to 150 mg, from 40 mg to 100 mg, from 45 mg to 75 mg, from 50 mg to 70 mg, from 55 mg to 65 mg, from 58 mg to 62 mg; e.g., about 60 mg). In some non-limiting embodiments, the dose is from about 12 mg to about 48 mg (e.g., from about 12 mg to about 42 mg, from about 12 mg to about 36 mg, from about 12 mg to about 30 mg, from about 18 mg to about 48 mg, from about 18 mg to about 42 mg, from about 24 mg to about 42 mg, from about 27 mg to about 42 mg, from about 24 mg to about 36 mg, from about 27 mg to about 33 mg, from about 28 mg to about 32 mg; e.g., about 24 mg, about 27 mg, about 30 mg, about 33 mg, or about 36 mg).


In certain non-limiting embodiments, the per day dosage for administration to a subject is in a range of 1 mg/kg to 25 mg/kg, or 2 mg/kg to 24 mg/kg, or 3 mg/kg to 22 mg/kg, or 4 mg/kg to 20 mg/kg, or 5 mg/kg to 17.5 mg/kg, or 5 mg/kg to 15 mg/kg, or 5 mg/kg to 12.5 mg/kg, or 6 mg/kg to 25 mg/kg, or 6 mg/kg to 20 mg/kg, or 6 mg/kg to 15 mg/kg, or 6 mg/kg to 12 mg/kg.


In certain non-limiting embodiments, the amount of the active agent delivered to the subject per dose is in a range of about 100 mg to about 1000 mg, or about 200 mg to about 1000 mg, or about 300 mg to about 1000 mg, or about 400 mg to about 1000 mg, or about 250 mg to about 900 mg, or about 400 mg to about 900 mg, or about 400 mg to about 850 mg, or about 400 mg to about 800 mg, or about 400 mg to about 700 mg, or about 420 mg to about 840 mg, or about 500 mg to about 650 mg, or about 500 mg to about 1000 mg, or about 500 mg to about 900 mg, or about 500 mg to about 850 mg, or about 500 mg to about 800 mg, or about 500 mg to about 700 mg. The total dose to be delivered can be provided in a single capsule, tablet, injection, or bolus (or other dosage form), or in multiple capsules, tablets, injections, or boluses (or other dosage forms).


In at least certain non-limiting embodiments, when the active agent is provided in the form of a capsule or tablet, the capsule or tablet should disintegrate within about 5-10 minutes, and in the gastrointestinal (GI) tract the active agent should dissolve in about 30 minutes.


In at least certain non-limiting embodiments, once the active agent is absorbed from the GI tract, it should produce a (therapeutic) concentration of at least 1.7 µg/ml, or at least 4 mM, in the target tissue. In at least certain non-limiting embodiments, the dosage form is designed to provide a therapeutic concentration of the active agent in the subject for at least about 12 to 24 hours, when the active agent is administered once a day.


The NCs presently disclosed have enhanced shelf-life stability. Nanocrystals prepared by the top-down method are likely to have amorphous parts, resulting from the heat produced while micronizing or milling the material, or exhibit stronger particulate interforces as a result of the charges produced by the micronization or milling. Therefore, they require specialized ingredients to be stabilized. In contrast, the NCs produced with the methods of the present disclosure are 100% crystalline and have a longer shelf life and do not require special excipients to preserve the stability of the drug in the nanocrystal.


Various embodiments of the present disclosure will be more readily understood by reference to the following examples and description, which as noted above are included merely for purposes of illustration of certain aspects and embodiments of the present disclosure, and are not intended to be limiting. The following detailed examples and methods which describe various compositions of the present disclosure and are to be construed, as noted above, only as illustrative, and not limitations of the disclosure in any way whatsoever.


EXAMPLES
Example 1: SHetA2 - PRIMA-1/PRIMA-1MET Drug Combinations
Methods
Cell Lines, Plasmids and Drugs

Ovarian cancer cells lines A2780 (RRID:CVCL_0134), OV-90 (RRID:CVCL_3768), SKOV3 (RRID:CVCL_0532), COV362 (RRID:CVCL_2420), OVSAHO (RRID:CVCL_3114), Caov3 (RRID:CVCL_0201), and OVCAR4 (RRID:CVCL_1627) (all from American Type Culture Collection/ATCC Manassas, VA, USA) were cultured in RPMI1640 medium containing 10% FBS and 1X antibiotic/antimycotic. The MESOV/GFP-luc (RRID:CVCL_CZ92) cell line (gift from Dr. Francois Moisan, Stanford University) (MESOV) was cultured in DMEM-high glucose medium with 10% FBS. SKOV3 cells stably transfected with mutant p53 (R248W, R273H) or parent vector were provided by Jeremy Chien, PhD (University of Kansas Medical Center). Human fallopian tube secretory epithelial cells (hFTSECs) were isolated from fallopian tube fimbriae surgical specimens under an IRB approved protocol. All cell lines were authenticated by autosomal short tandem repeat (STR) profiles determination within three years and comparison with reference databases by the University of Arizona Genetics Core and IDEXX BioAnalytics (Columbia, MO, USA). Experiments were performed with cell lines free of mycoplasma contamination. Cultures were transfected with plasmids HSPA9 pLX304 or pLKO-p53-shRNA-427, PG13-Luc (Addgene, Watertown, MA) and treated with SHetA2 and PRIMA-1 (National Cancer Institute PREVENT Program), and PRIMA-1MET (Cayman Chemicals, Michigan, USA) dissolved in dimethyl sulfoxide (DMSO). Caspase 3 inhibitor Z-DEVD-FMK and N-acetyl-L-Cysteine (Cayman Chemicals, Michigan, USA) were used to study mechanism of drug action.


TP53 Mutation Identification

Exome sequencing data from 429 TCGA ovarian cancer cases was analyzed for genome-wide mutations. The genomic coordinates of these mutations were converted from human genome build hg18 to hg19 using the UCSC Genome Browser liftOver tool (http://genome.ucsc.edu/cgi-bin/hgLiftOver). A total of 405 high-confidence mutations were located within the transcript locus of p53. Mutations were assigned to exons of ENSEMBL p53 transcripts on the basis of their location and predicted mutation effect.


Mortalin Expression in Ovarian Tissue

The Gynecologic Oncology Group (GOG) Disease Progression Tissue Microarray (TMA) was stained for mortalin using Abcam rabbit polyclonal antibody ab53098. Prior to staining the TMA, antibody dilutions and other immunohistochemical staining conditions were optimized on HGSOC sections using no primary antibody as negative controls. Heat-induced epitope retrieval was performed on formalin-fixed, paraffin-embedded TMA sections using a pressurized decloaking chamber (Biocare Medical, Pacheco, CA, USA) in citrate buffer (pH 6.0) at 99° C. for 18 min and then in 3% hydrogen peroxide at room temperature for 20 min. After incubation with primary antibody, positive signal was developed with a rabbit polymer-horseradish peroxidase secondary detection kit (Biocare Medical, Pacheco, CA, USA) and developed diaminobenzidine (Sigma, St. Louis, MO, USA). Digitized images of IHC-stained slides were generated and evaluated for mortalin positive staining intensities using the Positive Pixel Count algorithm software (Aperio Technologies, Inc., Vista, CA, USA). Positivity scores (total number of positive pixels divided by total number of pixels) of the mortalin staining was derived separately for epithelial and stromal cells by tuning a Genie algorithm to differentiate between these two cell types on mortalin-stained ovarian tissues. Marked-up images of the staining intensity measurements generated by the Aperio imaging software were reviewed manually and individual cores that exhibited artifacts, lack of tumor on the section, or inaccurate categorization of stromal versus epithelial cells were eliminated from the analysis. Each patient was represented on the slide by one to three sections. Individual cores that exhibited >10 fold difference from the two other cores from the same patient were eliminated from the averages.


Subcellular Fraction Enrichment

Cytoplasmic and nuclear protein fractions were isolated from cultures using NE-PER Nuclear and Cytoplasmic Extraction Reagents (ThermoFisher, Waltham, MA, USA) and the Mitochondria/Cytosol Fractionation Kit (ThermoFisher, Waltham, MA, USA) and protein concentrations determined with BCA reagent (ThermoFisher, Waltham, MA, USA).


Western Blots

Proteins were extracted from cell cultures using M-PER mammalian protein extraction reagent (ThermoFisher, Waltham, MA, USA) and volumes corresponding to 30 µg determined with BCA reagent (ThermoFisher, Waltham, MA, USA) were electrophoresed into a SDS-PAGE gel and transferred to PVDF membranes. Membranes were blocked (5% dry skim milk powder in TBST buffer) for at least 1 h or overnight, washed thrice with TBST and then incubated with primary antibody: p53 DO-1, β-actin, histone H3, GAPDH (Santa Cruz Biotechnology, Dallas, TX, USA) and mortalin, Tom20, V5 tag antibodies, p21, PARP, SLC7A11, caspase 3 (Cell Signaling Technology, Danvers, MA, USA) for at least 1 h or overnight. After three washes, membranes were incubated with anti-mouse HRP conjugated (Santa Cruz Biotechnology, Dallas, TX, USA) or anti-Rabbit HRP conjugated (Cell Signaling Technology, Danvers, MA, USA) for 40 min. Membranes then were washed and developed with ECL reagents (BioRad, Hercules, CA, USA). Bands were imaged and quantified (ChemiDoc imaging system with Image Lab Software/BioRad).


Immunofluorescence

Chamber slides (Lab-Tek II) were seeded with 1 × 104 cells in 1 ml medium and incubated up to 50-70% confluence. Cells treated with SHetA2 or an equivalent volume of DMSO were fixed in 2% paraformaldehyde in PBS, pH 7.4 for 30 min. at room temperature, washed with PBS three times and then permeabilized with 0.5% Triton X-100 in PBS, 7.4 for 5 minutes. After three PBS washes, cells were incubated in PBS containing 0.1% BSA in PBS for 30 min. at 37° C. and then with p53 DO-1 antibody for 1h at 37° C. After three PBS washes, cells were incubated with the secondary antibody tagged with fluorochrome (Alexa fluor 488 or 647) for 1 h at 37° C. and washed thrice in PBS. Nuclei were stained with 300 nM DAPI for 7 min at room temperature and cells were washed thrice in PBS. Cells were covered in Prolong Gold mounting medium (Life Technology, Carlsbad, CA, USA) and 1 mm thick cover glasses and left to dry overnight at room temperature and then imaged with a confocal microscope Leica SP2 (Leica, Wetzlar, Germany) at 63X.


MTT Cytotoxicity Assay

Cells plated at 2000 cells/well in 96 well microtiter plates were incubated for 36-48 h and treated with a range of doses and combinations of SHetA2 and PRIMA-1 or PRIMA-1MET, or the same volume of DMSO solvent for 72 h of incubation. The percentage of growth for single drug treatments or fold effect (1 - ratio of treated/untreated growth) was measured using the CellTiter 96 AQueous Non-Radioactive Cell Proliferation Assay (Promega, Madison, WI, USA) and the BioTeK SYNERGY H1 (BioTeK, Winooski, VT, USA). Each experiment was performed in triplicate and repeated 2-3 times.


Transfection of Cells

1×106 cells were electroporated with plasmid (5-10 µg) using the Neon transfection unit (Life technology, Waltham, MA) at 1170 V, 30 width and 2 pulses. Transfected cells were seeded onto 6-well and 96-well plates, and incubated for 36-48 h prior to evaluation by Western blot and MTT assays, respectively. A stable cell line of A2780 cells transfected with pLKO-p53-shRNA-427 was generated by transfecting A2780 cells using the electroporation conditions above. Transfected cells were titrated for the optimum puromycin concentration needed for the selection, which was determined to be 20 µg. Transfected cells were grown under puromycin pressure for 2 weeks and after that continued to grow in growth medium without puromycin. The puromycin-resistant cell line was evaluated for p53 knockdown using Western blot.


P53 Luciferase Reporter Assay

To determine the transcriptional activity of p53, MESOV cells were transfected with PG13-Luc plasmids. Approximately 5,000 transfected cells were seeded on to each well of 96-well plate and incubated overnight. These cells were treated with either control solvent, 5 µM SHetA2 or 60 µM PRIMA-1MET for 24 h. Luciferase activity of the reporter was measured with the One-glo luciferase reporter assay kit (Promega, Madison, WI, USA) and a Biotek Synergy H1 plate reader (BioteK, Winooski, VT, USA). The MTT assay was performed in parallel cultures and used to normalize the p53 reporter activity to the amount of viable cells.


Drug Interactions

The potencies (IC50′s) of SHetA2 or PRIMA-1 were determined using GraphPad Prism 6 software analysis of average MTT Optical Density vs drug dose. The drugs were mixed at a 1:1 ratio of their IC50 concentrations and a series of 2-fold dilutions evaluated in the MTT assay. Single drug treatments at doses surrounding the IC50 values were evaluated parallel. All treatments were performed in triplicate and averages used to draw isobolograms and calculate combination indices (CI) and dose-reduction indices (DRIs) using CompuSyn Software according to the Chou and Talalay method (op cit). All experiments were repeated at least twice for each cell line to confirm results.


Quantitative Reverse Transcriptase Polymerase Chain Reaction (qRT-PCR)

SKOV3 parental and SKOV3 R273H p53 mutant cells were grown to 80% confluency in a 100 mm tissue culture dish and then treated with SHetA2 10 µM or PRIMA-1 20 µM or both drugs combined for 16 h after which cells were harvested and RNA was isolated using RNeasy Mini Kit (QIAGEN). RNA quality and concentration was evaluated by measuring the OD260/280 ratio. The cDNA was synthesized from 1 µg RNA using RT2 First Strand Kit (Qiagen, Hilden, Germany) and then Real-time PCR was performed by using RT2 SYBR Green master mix (GeneCopoeia, Rockville, MD, USA) and the BioRad CFX96 Real Time PCR detection system (Bio Rad, Winooski, VT, USA). The real time expression values were analyzed by calculating ΔΔCT and mean fold change (2^- (ΔΔCT)) values.


Assays for Reactive Oxygen Species (ROS) and ATP

The ROS-Glo H2O2 Assay and CellTiter-Glo 2.0 Assay (Promega, Madison, WI, USA) kits were used to measure ROS and ATP, respectively, in cells cultured on 96-well plates with various combinations of SHetA2 (5 µM, 10 µM) and PRIMA-1 or PRIMA-1MET (25 µM) combinations, and the luminescence endpoints were measured with a BioTeK SYNERGY H1. The ROS assay luminescence endpoints were normalized to total viable cells using the MTT assay. For ROS to ATP ratio determination, the ROS luminescence endpoint was normalized to total protein content (µg/µl) measured with the BCA reagent in parallel wells, and the ATP concentrations were derived from a standard curve and normalized to total protein content. All assays were performed in triplicate and repeated three times.


Animal Model

Four-week-old female athymic nude mice (NCr-Foxn1nu) were obtained from Taconic Biosciences under an approved Institutional Animal Care and Use Committee (IACUC) protocol. Mice were acclimatized for two weeks, fed with a standard diet and ear tagged during the acclimatization period. The timing to tumor development after intraperitoneal injection with various amounts of MESOV cells was determined in a pilot study prior. In the intervention experiment, 6-week-old mice were injected with 20 million MESOV human ovarian cancer cells in their intraperitoneal region. Drug treatment was initiated 4 weeks after the injection of cancer cells. Mice were randomized to four treatment groups before the start of treatment based on their body weight. There was no significant (one-way ANOVA, p >0.999) difference in body weight between groups at the start of treatment. Twelve mice were allocated to each group based on the power analysis performed while designing experiment. The sample size was determined to provide over 80% power to detect a 915 mm3 difference in tumor volume between PRIMA-1MET treatment alone and the drug combination treatment using a two-sided 0.05 level t-test. SHetA2 was administered by gavage at 60 mg/kg in 30% Kolliphor HS15 (Sigma). Controls were gavaged with Kolliphor HS15 (30% in water). PRIMA-1MET (10 mg/kg) was dissolved in phosphate buffered saline and injected into the peritoneum of mice every other day. Initially SHetA2 dosing was administered every day for two weeks in SHetA2 alone and drug combination groups, and then switched to every other day for the next three weeks due to the stress caused to the mice by daily gavage. Oral gavaging was performed with sterile disposable plastic feeding tubes 20 ga × 38 mm (Instech Laboratories, Inc., Plymouth Meeting, PA, USA). Three mice in the drug combination group died after two weeks of drug treatment due to gavaging incidents. Body weights were recorded and animal health was monitored weekly. After the end of drug intervention period, all mice were euthanized by CO2 asphyxiation followed by exsanguination, their tumors and other vital organs were collected for further analysis. After measuring the total tumor weight, a portion of the tumors were snap frozen in liquid nitrogen and another portion was formalin fixed. The formalin fixed tissue samples were paraffin embedded, sectioned and stained with hematoxylin and eosins for histology studies. An experienced pathologist reviewed blinded histology slides for kidney and liver toxicity.


Statistics

For tissue culture studies, the normality of data and statistical significance were analyzed using Prism 8 Software (GraphPad, San Diego, CA, USA). Comparison of drug effects in the presence and absence of p53 knockdown or mortalin overexpression were done using T-tests corrected for multiple comparisons using the Holm-Sidak method. Comparison of drug combinations compared to single drug treatments were done using a two-way ANOVA with a Tukey’s multiple comparison test. P values < 0.05 were considered significant.


Presence or absence of tumor at the end of the treatment period was used as the primary outcome measure. A binary variable (no tumor) was created which equaled 1 if the tumor weight was zero and equaled 0 otherwise. Two by two logistic regression was used to compare the four treatment groups (Untreated control, SHetA2, PRIMA1MET, SHetA2+PRIMA1MET combination) in the study. Mice in the first 3 groups had complete data, while 3 mice in the combo group died early and had zero tumor weights. To assess the synergistic effects of SHetA2 and PRIMA1MET, a logistic regression model was used to analyze the no tumor variable with two factors SHetA2 (indicating whether SHetA2 was part of the treatment) and PRIMA1MET (indicating whether PRIMA1MET was part of the treatment) and their interaction. The interaction coefficient was not included in the model because it was not significant, which resulted in an additive model. Statistical Analysis (SAS 9.4) was used to perform the analysis. All of the tumors found and excised during necropsy were weighed simultaneously to obtain the tumor burden, which was considered a continuous variable. Tumor burdens between the different treatment groups were compared by the Kruskal-Wallis test using Prism 8.0 (GraphPad Software, San Diego, CA, USA).


Results
Alterations of P53 and Mortalin in Ovarian Cancer Specimens

Various missense p53 mutations commonly present in cancer exert different functions that could cause dissimilar consequences on responses of tumors to therapy, and therefore have potential to predict treatment response when evaluated individually. To evaluate the profile of TP53 mutations in TCGA HGSOC data, Ensembl exons of p53 genomic regions corresponding to various domains of the p53 protein were evaluated individually (See FIG. 1A of Provisional Application Serial No. 62/730,345, filed Sept. 12, 2018). TP53 exon mutations in TCGA HGSOC data occurred at a 94.4% (405/429) frequency. The majority (58%, 234/405) were missense mutations within the DNA binding domain (aa 102-292). Another 3% (12/405) were in regions outside of the DNA binding domain. The types of mutations known, or likely, to result in loss of full-length p53 protein expression from one allele (frameshift deletions and insertions, nonsense mutations and splice site mutations) totaled 39% (157/405). The most frequently mutated amino acids in p53 protein were R248, R273, R175, G245, R249 and R282. HSPA9 (mortalin) gene mutations were infrequent.


The status of the SHetA2 drug target mortalin in a patient’s tumor is also likely to affect treatment outcome. The present analysis of TCGA data found no association of mortalin mutations with ovarian cancer. IHC analysis of mortalin expression in a series of serous benign, borderline and cancerous tissues on a TMA demonstrated punctate expression of mortalin in both epithelial and cancer cells consistent with its primary localization in mitochondria (FIG. 1). Mortalin positivity scores in epithelial and stromal cells significantly increased in a step-wise manner from benign to borderline to cancerous tissues (FIGS. 2 and 3, respectively; Kruskal-Wallis test p = 0.0282 for epithelial cells; p = 0.0002 for stromal cells).


Effects of SHetA2 on P53 Cellular Localization

We predicted that SHetA2 disruption of mortalin/p53 complexes would cause p53 to translocate from the cytoplasm where it is sequestered by mortalin to accumulate in its nuclear and mitochondrial sites of action. Immunofluorescent imaging confirmed SHetA2 dose-dependent elevation of both wild type and mutant p53 in nuclei of ovarian cancer cell lines, but not in hFTSECs (FIG. 4). Very little p53 was detectable in the cytoplasm likely due to masking of the epitope by binding proteins and the short p53 half-life. Western blots demonstrated SHetA2 dose-dependent decreases in cytoplasmic fractions and increases in nuclear (FIGS. 5-6) and mitochondrial (FIGS. 7-8) fractions of the A2780 wild type p53 and OV90 S215R or OVCAR4 m-p53 cell lines. SHetA2 caused minimal effects on p53 cellular localization in human fallopian tube secretory epithelial cells (hFTSECs) (FIGS. 9-10). In contrast to p53, mortalin cellular or nuclear levels were not affected by SHetA2 treatment in these cell types except for one instance of nuclear accumulation in A2780 cells treated with 5 µM SHetA2 (FIGS. 5,6,9).


Effects of SHetA2 and PRIMA-1 as Single Agents

shRNA knockdown of p53 reduced SHetA2 potency in A2780 cells from a 2.8 µM to 4.8 µM IC50 (FIG. 11). Overexpression of mortalin in A2780 cells also reduced SHetA2-induced cytotoxicity (FIG. 12), likely due to mortalin sequestering SHetA2 away from mortalin/p53 complexes. Ovarian cancer cell lines expressing m-p53 were more resistant to SHetA2 in comparison to the wild type p53 A2780 cell line, while hFTSECs were the least sensitive (FIG. 13). SKOV3 p53 null cells permanently transfected to express m-p53 exhibited increased PRIMA-1 sensitivity in comparison to PRIMA-1-resistant parental SKOV-3 cells to different extents depending on the mutant (R248W: 19.98 µM IC50; R273H:IC50 39.96 µM IC50) (FIG. 14). PRIMA-1MET reactivation of R282W m-p53 in MESOV was observed using a p53 reporter plasmid (FIG. 15).


Effects of SHetA2 and PRIMA-1/PRIMA-1MET in combination

SHetA2 and PRIMA-1 showed synergism in ovarian cancer cells harboring wild type p53 or m-p53, additive interaction in Caov3 p53 null cells and antagonism in hFTSECs (FIGS. 16-20 and Table 12). SHetA2 and PRIMA-1MET also showed synergism in m-p53 MESOV cells and antagonism in hFTSECs (FIGS. 21-22 and Table 2). DRIs of SHetA2 and PRIMA-1/PRIMA-1MET combinations indicated that the combination allows lower doses of each drug to be used without reducing activity (Table 13). For example, for the cell line A2780, in the case of when SHetA2 and PRIMA-1 are used in combination, 3.84 times less SHetA2, and 2.12 times less PRIMA-1 can be used to achieve the same IC50 as that obtained by using either drug alone.





TABLE 12









Combination index values at various Effective Doses (EDs) as calculated by CompuSyn using the Chou-Talalay method


Combinations
Combination index (CI)* at
Combination analysis result


SHetA2+ PRIMA-1
ED50
ED75
ED90
ED95





A2780 (WT p53)
0.73
0.74
0.74
0.75
Synergism


MESOV (R282Wp53)
0.81
0.75
0.73
0.73
Synergism


SKOV3 R248W p53
0.96
0.75
0.59
0.50
Synergism


CaOV3 (Null p53)
1.04
1.03
1.04
1.05
Additive


FTSECs
2.5
1.57
0.97
0.71
Antagonism












SHetA2 + PRIMA-1MET




MESOV (R282Wp53)
1.01
0.82
0.66
0.57
Synergism


FTSECs
1.70
1.86
2.05
2.21
Antagonism


*CI<1, =1 and > 1 indicates synergism, additive effect, and antagonism, respectively.









TABLE 13








Dose reduction indices of SHetA2 and PRIMA-1/PRIMA-1MET drug combinations in various cell lines


Drug Combinations/ Cell lines
Dose-reduction index (DRI)* (SHetA2/PRIMA-1 or SHetA2/PRIMA-1MET) at


SHetA2+ PRIMA-1
IC50
IC75
IC90
IC95




A2780 (WT p53)
3.84/2.12
3.72/2.13
3.6/2.14
3.53/2.15


MESOV (R282Wp53)
1.79/3.94
2.21/3.27
2.73/2.71
3.15/2.38


SKOV3 R248W p53
2.48/1.79
3.03/2.38
3.70/3.15
4.24/3.81


CaOV3 (Null p53)
2.48/1.56
2.74/1.48
3.01/1.40
3.22/1.35


FTSECs
1.62/0.52
2.37/0.86
3.48/1.44
4.50/2.03











SHetA2 + PRIMA-1MET




MESOV (R282Wp53)
1.84/2.10
2.17/2.76
2.56/3.64
2.87/4.38


FTSECs
2.30/0.78
2.59/0.67
2.90/0.58
3.14/0.52


*Folds of dose reduction when compared with amount of each drug alone.






Evaluation of SHetA2 and PRIMA-1 Synergy Mechanism

The TP21 gene was evaluated as a marker of p53 transcriptional activity. SHetA2 induced TP21 transcription in p53 null and m-p53 -expressing SKOV3 cells (FIG. 23), and increased p21 protein in all cell lines evaluated regardless of p53 status (FIGS. 24-27). PRIMA-1 did not induce TP21 transcription in m-p53-expressing SKOV3 cells (FIG. 23) and only induced p21 protein expression in A2780 (wt p53) (FIG. 24). PRIMA-1 prevented SHetA2 induction of p21 only in the presence of R248W m-p53, but did not prevent PARP-1 cleavage as an indicator of apoptosis (FIGS. 28-31).


SHetA2 also induced SLC7A11, a cysteine/glutamate antiporter shown to be inversely associated with m-p53 expression and PRIMA-1MET sensitivity by others and confirmed in this study (compare FIGS. 14 and 24-32). The extents of SLC7A11 repression and ROS generation were associated with PRIMA-1 sensitivity (compare FIGS. 14 and 33-36). Involvement of ROS in the synergy mechanism was suggested by higher levels of ROS generation in SKOV3 cells treated with SHetA2 and PRIMA-1 in comparison to single drug treatments, which occurred in an additive manner in p53 null cells and in a synergistic manner in m-p53 expressing cells (FIGS. 33-35). Levels of cellular ATP were decreased as levels of ROS were increased in SKOV3 p-53 null and m-p53 cells treated with SHetA2 and PRIMA-1 (compare FIGS. 14 and 36). ATP levels were also reduced in association with increased ROS by combined treatment with SHetA2 and PRIMA-1MET in A2780 wild type p53 cells, and this effect was antagonized by p53 shRNA (FIGS. 37-38). Involvement of apoptosis in the synergy mechanism was verified by caspase 3-inhibitor, Z-DEVD-FMK, reduction of caspase 3 and PARP-1 cleavage across all cell lines treated with SHetA2 and PRIMA-1MET (FIGS. 28-31).


Evaluation of SHetA2 and PRIMA-1MET in a Model of Secondary Chemoprevention

To model maintenance therapy for ovarian cancer, effects of SHetA2 and PRIMA-1MET on establishment of tumors from MESOV cells injected into the peritoneum of immunocompromised mice were evaluated. Treatment was initiated 28 days post-injection and continued for 38 days. Treated mice appeared normal, however the average body weights of the PRIMA-1MET group were significantly higher than the mock group (FIG. 39). This was attributed to reduced gavage-associated stress and development of ascites in the PRIMA-1MET group (FIG. 39). At necropsy, all control group mice had solid tumor nodules with variable numbers and sizes (FIG. 40). Tumors were commonly observed on uterine horns, intestine, chest cavity, stomach, near the spleen and below the kidney and pancreas. Tumor-free rates were 0% in the untreated controls, 25% in the PRIMA1MET treatment group, 42% in the SHetA2 treatment group, and 67% in the combination treatment group (FIG. 41). Logistic regression indicated that the interaction between SHetA2 and PRIMA1MET was not significantly synergistic (p=0.973). A subsequent additive model showed that both SHetA2 (p=0.004, OR=10.384, 95% CI: 2.158, 48.965) and PRIMA1MET (p=0.048, OR=4.464, 95% CI: 1.014, 19.655) significantly prevented tumor development, and that these two drugs acted additively in preventing tumor establishment.


Histologic analysis of kidney and liver collected upon necropsy showed no evidence of toxicity (FIG. 42). Western blot analysis of tumors confirmed expression of Pax8, a marker of high-grade serous ovarian cancer (FIG. 43A). There was no association of p53 expression with treatment (FIG. 43A). SLC7A11 was increased significantly in the drug combination group compared to control group (FIG. 43B).


These results provide justification for targeting m-p53 and mortalin in development of ovarian cancer therapy. TCGA data analysis revealed a 58% m-p53 rate in HGSOC. TMA analysis demonstrated elevated mortalin expression in serous cancer. Two drugs, SHetA2 and PRIMA-1MET, which wield complementary mechanisms targeted at m-p53 and mortalin, exerted synergistic cytotoxicity against ovarian cancer cell lines expressing m-p53 and additive activity in a p53 null cell line. The antagonism observed in hFTSEC cells suggests that this combination will not be toxic in clinical trials. The significant in vivo prevention of tumor establishment by each drug alone and the additive activity when used in combination validate the tissue culture results.


We predicted that SHetA2 would increase p53 activity because it releases p53 from binding to mortalin, and mortalin can sequester p53 in the cytoplasm away from its apoptosis sites of action in stressed cancer cells. In the present work, SHetA2 caused p53 accumulation in nuclei and mitochondria in ovarian cancer cells, but not in hFTSECs. This differential effect may explain the lack of observed toxicity for SHetA2. Targeting mortalin/p53 has limitations in HGSOC, due to high m-p53 frequency and our observation that cell lines expressing endogenous or exogenous m-p53 exhibited increased SHetA2 resistance compared to wild type or p53 null cell lines.


We hypothesized that a combination of a heteroarotinoid such as SHetA2 with a p53 reactivator drug would be synergistic in m-p53 cell lines, because SHetA2 would increase the amount of p53 available to induce apoptosis while the p53-reactivator would restore apoptosis function to released m-p53. The results support this hypothesis in that SHetA2 and PRIMA-1 or PRIMA-1MET acted synergistically in cytotoxicity assays of ovarian cancer cell lines expressing m-p53, but only additively in p53 null lines, and antagonistically in hFTSECs. A first-in-human clinical trial of PRIMA-1MET infusion in cancer patients found that PRIMA-1MET reached its target in tumors and was relatively safe with fully reversible central nervous system-related side effects. The DRI’s generated in the present work indicates that these side effects can be reduced or eliminated by combination treatment with SHetA2 allowing lower doses of PRIMA-1MET to be used.


The present work indicates that SHetA2 and PRIMA-1/PRIMA-1MET drug combinations induce apoptotic cell death by pushing cells past the “point of no return” to a ratio of ROS/ATP that is increased beyond a point at which recovery is impossible as defined by the Nomenclature Committee on Cell Death. Both wild type and m-p53 proteins appear to be involved in this mechanism as ovarian cancer cells expressing wild type p53, or endogenous or exogenous m-p53, exhibited greater ROS/ATP ratio elevation upon drug combination treatment compared to cells that were p53 null or had reduced p53. SLC7A11 levels were inversely associated with PRIMA-1 sensitivity, but not SHetA2/PRIMA-1 sensitivity, possibly due to the SHetA2 induction of SLC7A11 expression. In oesophageal cancer, m-p53 increased PRIMA-1MET sensitivity, m-p53/NRF2 complexes repressed SLC7A11 expression and SLC7A11 overexpression decreased, while knockdown increased, PRIMA-1MET sensitivity in m-p53 cells.


Current ovarian cancer maintenance therapy is limited by toxicity and no proven impact on overall survival. To study a maintenance strategy targeted at missense mutant p53, we hypothesized that release of mutant p53 from mortalin inhibition by the SHetA2 drug combined with reactivation of mutant p53 with the PRIMA-1MET drug inhibits growth and tumor establishment synergistically in a mutant-p53 dependent manner. TCGA data and serous ovarian tumors were evaluated for TP53 and HSPA9/mortalin status. SHetA2 and PRIMA-1MET were tested in ovarian cancer cell lines and fallopian tube secretory epithelial cells using isobolograms, fluorescent cytometry, Western blots and ELISAs. Drugs were administered to mice after peritoneal injection of MESOV mutant p53 ovarian cancer cells and prior to tumor establishment, which was evaluated by logistic regression. Fifty-eight percent of TP53 mutations were missense and there were no mortalin mutations in TCGA high-grade serous ovarian cancers. Mortalin levels were sequentially increased in serous benign, borderline and carcinoma tumors. SHetA2 caused p53 nuclear and mitochondrial accumulation in cancer, but not in healthy, cells. Endogenous or exogenous mutant p53 increased SHetA2 resistance. PRIMA-1MET decreased this resistance and interacted synergistically with SHetA2 in mutant and wild type p53-expressing cell lines in association with elevated ROS/ATP ratios. Tumor-free rates in animals were 0% (controls), 25% (PRIMA1MET), 42% (SHetA2) and 67% (combination). SHetA2 (p=0.004) and PRIMA1MET (p=0.048) functioned additively in preventing tumor development with no observed toxicity.


Example 2: SHetA2 - Palbociclib Drug Combination

CDK 4/6 inhibitors (e.g., Palbociclib, Ribociclib, Abemaciclib, ON123300) are structurally-related drugs currently FDA approved for treatment of cancer. As a demonstration that SHetA2 works synergistically with the CDK 4/6 group of drugs, SHetA2 was tested for its ability to interact synergistically with palbociclib.


Palbociclib, an FDA approved anticancer drug for treatment of breast cancer, was evaluated in combination with SHetA2 because they inhibit cyclin D1/CDK4/6 complex-driven cancer cell growth through complementary mechanisms. Palbociclib binds and inhibits the CDK4/6 kinase activity. SHetA2 disrupts mortalin complexes and mortalin is known to bind cyclin D1. Another SHetA2 binding target, Hsc70, stabilizes the cyclin D1/CDK4/6 complex. Furthermore, SHetA2 causes degradation of cyclin D1 in cell lines, and in animal models in association with tumor reduction.


Strong synergy was observed between SHetA2 and Palbociclib when used to treat three human cervical cell lines (SiHa, Caski, C33a) (FIGS. 44-46 and Table 14). Combination Indices (CI’s) were derived to determine the level of synergy using the Chow and Talalay method. All of the cell lines exhibited CI’s indicative of strong synergy at effective doses (EDs) causing 95% (ED95), 90% (ED90) and 75% (ED75) cell reduction (Tables 14 and 15). The CI’s at the ED50 were indicative of strong synergy for the SiHa and Caski cell lines, and synergy for the C33a cell line (Tables 14 and 15). The Dose Reduction Indices (DRIs) were derived to determine the level of reduction for one drug that could be allowed by addition of the second drug at various fold effects (Fas) of the combination treatment (Table 16).





TABLE 14








Combination Index for Various Effective Doses (ED) of SHetA2 and Palbociclib Cervical Cancer Cell Lines


Cell Line
ED50
ED75
ED90
ED95




SiHa
0.205
0.113
0.093
0.089


Caski
0.218
0.238
0.296
0.361


C33a
0.512
0.222
0.195
0.221









TABLE 15





Interpretation of Combination Index (CI)


CI Range
Interpretation




<0.1
Very Strong Synergism


0.1 - 0.3
Strong Synergism


0.3 - 0.7
Synergism


0.7 - 0.85
Moderate Synergism


0.85 - 0.90
Slight Synergism


0.90 - 1.10
Nearly Additive


1.10 - 1.20
Slight Antagonism


1.20 - 1.45
Moderate Antagonism


1.45 - 3.3
Antagonism


3.3 - 10
Strong Antagonism









TABLE 16








Dose Reduction Index (DRI) of SHetA2-Palbociclib Combinations at Various Fold Effects (Fas) in Cervical Cancer


Fold Effect (Fa)
SHetA2 Dose
Palbociclib Dose
SHetA2 DRI
Palbociclib DRI




0.05
5.44
7.16
8.76
9.6


0.75
8.23
24.03
5.92
14.42


0.90
12.43
80.72
4.003
21.66


0.95
16.47
184.00
3.068
28.57






Induction of cell death contributes to the synergistic effect in SiHa and C33A cells (FIGS. 47-48, respectively). An animal model validated the efficacy of SHetA2 and Palbociclib against SiHa human cervical cancer cell line xenografts (FIG. 49). The combination of SHetA2 and palbociclib exerted greater reduction of tumor growth in comparison to palbociclib alone (Repeated Measures ANOVA multiple comparison adjusted p value = 0.001 for comparison of the palbociclib and SHetA2 + palbociclib groups).


A combination treatment of SHetA2 and Palbociclib also has synergistic activity against endometrial cancer, as shown with two human endometrial cancer cell lines, Heclb (FIG. 50) and Ishikawa (FIG. 51). The indicated cell lines were treated with a dilution series of palbociclib, SHetA2 or 1:1 ratio of the half maximal inhibitory concentrations of palbociclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa).


Example 3: SHetA2 + Abemaciclib Drug Combinations

SHetA2 synergizes with abemaciclib against ovarian cancer spheroids. Evaluation of SHetA2+abemaciclib drug combinations in spheroids generated from ovarian cancer cell lines (OVCAR-8, OV-90, and ES2) demonstrates that that the combination therapy can be used for improved ovarian cancer maintenance therapy (FIG. 52).



FIGS. 53-54 show synergy between SHetA2 and abemaciclib in the human ovarian cancer cell lines MESOV and OC-90, respectively. The cell lines were treated with a dilution series of abemaciclib, SHetA2 or 1:1 ratio of the half maximal inhibitory concentrations of abemaciclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa).


SHetA2 Synergizes With Abemaciclib Against Endometrial Cancer.

A combination treatment of SHetA2 and abemaciclib also has synergistic activity against endometrial cancer, as shown with three human endometrial cancer cell lines, AN3CA (FIG. 55), Heclb (FIG. 56) and Ishikawa (FIG. 57). The indicated cell lines were treated with a dilution series of abemaciclib, SHetA2, or 1:1 ratio of the half maximal inhibitory concentrations of abemaciclib for 72 hrs. The MTT cytotoxicity assays was used to measure viable cells at the end of treatment and the results were interpreted using the Chou-Talalay Method using CompuSyn. The isobolograms show that the combination index indicated by the single symbol falls below the equivalent dose-effect line indicating synergy for each fold affect (Fa).


Example 4: SHetA2 + ON123300 Drug Combinations
SHetA2 Synergizes with ON123300 Against Ovarian Cancer Spheroids.

SHetA2 synergizes with ON123300 against ovarian cancer spheroids. Evaluation of SHetA2+ON123300 drug combinations in spheroids generated from ovarian cancer cell lines (OVCAR-8, OV-90, and ES2) demonstrates that that the combination therapy can be used for improved ovarian cancer maintenance therapy (FIG. 58).


SHetA2 is active against all cancers present in the NCI 60-cell line panel and also against cervical and esophageal cancer cell lines. Thus, it is reasonable to expect that the SHetA2 drug combinations with Azabicyclooctan-3-one derivatives, and CDK4/6 inhibitors will also be effective in these cancer types.


In accordance with the foregoing, the present disclosure is directed to, in at least certain embodiments:


Clause 1. A drug combination for use in a cancer treatment method in a subject in need of such therapy, wherein the cancer treatment comprises administering the drug combination to the subject, and the drug combination comprises a therapeutically-effective amount of a heteroarotinoid compound and a second compound selected from the group consisting of Azabicyclooctan-3-one derivatives, and CDK4/6 inhibitors, wherein the therapeutically-effective amount yields a synergistic effect compared to an effect of the heteroarotinoid alone and an effect of the second compound alone.


Clause 2. The drug combination of clause 1, wherein the cancer treatment results in at least one of cancer cell death, reduction of tumor size, reduction of tumor growth, inhibition of cancer metastases, and inhibition of tumor recurrence.


Clause 3. The drug combination of clause 1 or 2, wherein the heteroarotinoid is selected from the group consisting of SHetA2, SHetA3, SHetA4, SHetC2, SHetD3, SHetD4, SHetD5, SHet50, SHet65, SHet100, OHet72, NHet17, NHet86, and NHet90.


Clause 4. The drug combination of any one of clauses 1-3, wherein the second compound is an Azabicyclooctan-3-one derivative.


Clause 5. The drug combination of clause 4, wherein the Azabicyclooctan-3-one derivative is selected from PRIMA-1met and PRIMA-1.


Clause 6. The drug combination of any one of clauses 1-3, wherein the second compound is a CDK4/6 inhibitor.


Clause 7. The drug combination of clause 6, wherein the CDK4/6 inhibitor is selected from the group consisting of Palbociclib, Abemaciclib, Ribociclib, Palbociclib, Abemaciclib, Ribociclib, Narazaciclib (ON123300), Dalpiciclib, Dinaciclib, Milciclib, Seliciclib, Wang-4d, and Wang-4e.


Clause 8. The drug combination of claim 1, wherein the heteroarotinoid compound and a second compound are selected from the group consisting of the drug combinations in Tables 10-13.


Clause 9. The drug combination of any one of clauses 1-8, wherein the heteroarotinoid is administered in a dose range of 1 mg/kg to 100 mg/kg, and the second compound is administered in a dose range of of 1 mg/kg to 100 mg/kg.


Clause 10. The drug combination of any one of clauses 1-9, wherein the heteroarotinoid and the second compound are administered in a weight-to-weight ratio in a range of 50:1 to 1:50.


Clause 11. A drug combination comprising a heteroarotinoid, and a second compound selected from the group consisting of Azabicyclooctan-3-one derivatives and CDK4/6 inhibitors, wherein the drug combination provides for a synergistic effect against cancer cells compared to an effect of the heteroarotinoid alone and an effect of the second compound alone.


Clause 12. The drug combination of clause 11, wherein the heteroarotinoid and the second compound are combined in a single composition, or are present in a kit of parts comprising at least two separate compositions for sequential or simultaneous administration to a subject.


Clause 13. The drug combination of clause 11 or 12, wherein the inhibition of cancer cells results in at least one of cancer cell death, reduction of tumor size, reduction of tumor growth, inhibition of cancer metastases, and inhibition of tumor recurrence.


Clause 14. The drug combination of any one of clauses 11-13, wherein the heteroarotinoid is selected from SHetA2,, SHetA3, SHetA4, SHetC2, SHetD3, SHetD4, SHetD5, SHet50, SHet65, SHet100, OHet72, NHet17, NHet86, and NHet90.


Clause 15. The drug combination any one of clauses 11-14, wherein the second compound is an Azabicyclooctan-3-one derivative.


Clause 16. The drug combination of clause 15 wherein the Azabicyclooctan-3-one derivative is selected from PRIMA-1met and PRIMA-1.


Clause 17. The drug combination of any one of clauses 11-14, wherein the second compound is a CDK4/6 inhibitor.


Clause 18. The drug combination of clause 17, wherein the CDK4/6 inhibitor is selected from the group consisting of Palbociclib, Abemaciclib, Ribociclib, Narazaciclib (ON123300), Dalpiciclib, Dinaciclib, Milciclib, Seliciclib, Wang-4d, and Wang-4e..


Clause 19. The drug combination of any one of clauses 11-14, wherein the heteroarotinoid compound and a second compound are selected from the group consisting of the drug combinations in Tables 10-13.


Clause 20. The drug combination of any one of clauses 11-19, wherein the heteroarotinoid comprises a dosage in a range of 1 mg/kg to 100 mg/kg, and the second compound comprises a dosage in a range of 1 mg/kg to 100 mg/kg.


Clause 21. The drug combination of any one of clauses 11-20, wherein the heteroarotinoid and the second compound comprise a weight-to-weight ratio in a range of 50:1 to 1:50.


Clause 22. A kit or commercial package, comprising a hetertoarotinoid, and a second compound selected from the group consisting of Azabicyclooctan-3-one derivatives, and CDK4/6 inhibitors, wherein the hetertoarotinoid and the second compound when administered conjointly provide for a synergistic effect against cancer cells compared to an effect of the heteroarotinoid alone and an effect of the second compound alone.


Clause 23. The kit or commercial package of clause 22, wherein the inhibition of cancer cells results in at least one of cancer cell death, reduction of tumor size, reduction of tumor growth, inhibition of cancer metastases, and inhibition of tumor recurrence.


Clause 24. The kit or commercial package of clause 22 or 23, wherein the heteroarotinoid is selected from SHetA2, SHetA3, SHetA4, SHetC2, SHetD3, SHetD4, SHetD5, SHet50, SHet65, SHet100, OHet72, NHet17, NHet86, and NHet90.


Clause 25. The kit or commercial package of any one of clauses 22-24, wherein the second compound is an Azabicyclooctan-3-one derivative.


Clause 26. The kit or commercial package of clause 25, wherein the Azabicyclooctan-3-one derivative is selected from PRIMA-1met and PRIMA-1.


Clause 27. The kit or commercial package of any one of clauses 22-24, wherein the second compound is a CDK4/6 inhibitor.


Clause 28. The kit or commercial package of clause 27, wherein the CDK4/6 inhibitor is selected from the group consisting of Palbociclib, Abemaciclib, Ribociclib, Narazaciclib (ON123300), Dalpiciclib, Dinaciclib, Milciclib, Seliciclib, Wang-4d, and Wang-4e..


Clause 29. The kit or commercial package of claim 22, wherein the heteroarotinoid compound and a second compound are selected from the group consisting of the drug combinations in Tables 10-13.


Clause 30. The kit or commercial package of any one of clauses 22-29, wherein the heteroarotinoid comprises at least one dosage in a range of 1 mg/kg to 100 mg/kg, and the second compound comprises at least one dosage in a range of 1 mg/kg to 100 mg/kg.


Clause 31. The kit or commercial package of any one of clauses 22-30, comprising a dosage of the heteroarotinoid and the second compound having a weight-to-weight ratio in a range of 50:1 to 1:50.


While the present disclosure has been described in connection with certain embodiments so that aspects thereof may be more fully understood and appreciated, it is not intended that the present disclosure be limited to these particular embodiments. On the contrary, it is intended that all alternatives, modifications and equivalents are included within the scope of the present disclosure. Thus the examples described above, which include particular embodiments, will serve to illustrate the practice of the present disclosure, it being understood that the particulars shown are by way of example and for purposes of illustrative discussion of particular embodiments only and are presented in the cause of providing what is believed to be the most useful and readily understood description of procedures as well as of the principles and conceptual aspects of the presently disclosed methods and compositions. Changes may be made in the formulation of the various compositions described herein, the methods described herein or in the steps or the sequence of steps of the methods described herein without departing from the spirit and scope of the present disclosure.

Claims
  • 1. A cancer treatment method for treating a subject in need of such therapy, comprising: conjointly administering a therapeutically-effective amount of a heteroarotinoid compound and a second compound selected from the group consisting of an Azabicyclooctan-3-one derivative and a CDK4/6 inhibitor, wherein the therapeutically-effective amount yields a synergistic effect compared to an effect of the heteroarotinoid alone and an effect of the second compound alone.
  • 2. The cancer treatment method of claim 1, wherein the cancer treatment method results in at least one of reduction of tumor size, reduction of tumor growth, inhibition of cancer metastases, cancer cell death, and inhibition of tumor recurrence.
  • 3. The cancer treatment method of claim 1, wherein the heteroarotinoid is selected from SHetA2, SHetA3, SHetA4, SHetC2, SHetD3, SHetD4, SHetD5, SHet50, SHet65, SHet100, OHet72, NHet17, NHet86, and NHet90.
  • 4. The cancer treatment method of claim 1, wherein the second compound is an Azabicyclooctan-3-one derivative selected from PRIMA-1met and PRIMA-1.
  • 5. The cancer treatment method of claim 1, wherein the second compound is a CDK4/6 inhibitor.
  • 6. The cancer treatment method of claim 5, wherein the CDK4/6 inhibitor is selected from the group consisting of Palbociclib, Abemaciclib, Ribociclib, Narazaciclib (ON123300), Dalpiciclib, Dinaciclib, Milciclib, Seliciclib, Wang-4d, and Wang-4e..
  • 7. The cancer treatment method of claim 1, wherein the heteroarotinoid compound and the second compound are selected from the group consisting of the drug combinations in Tables 10-13.
  • 8. The cancer treatment method of claim 1, wherein the heteroarotinoid is administered in a dose range of 1 mg/kg to 100 mg/kg, and the second compound is administered in a dose range of of 1 mg/kg to 100 mg/kg.
  • 9. The cancer treatment method of claim 1, wherein the heteroarotinoid and the second compound are administered in a weight-to-weight ratio in a range of 50:1 to 1:50.
  • 10. A drug combination comprising a hetertoarotinoid, and a second compound selected from the group consisting of Azabicyclooctan-3-one derivatives, and CDK4/6 inhibitors, wherein the drug combination has a synergistic effect against cancer cells compared to an effect of the heteroarotinoid alone and an effect of the second compound alone.
  • 11. The drug combination of claim 10, wherein the heteroarotinoid and the second compound are combined in a single composition, or are present in a kit of parts comprising at least two separate compositions for sequential or simultaneous administration to a subject.
  • 12. The drug combination of claim 10, wherein the inhibition of cancer cells results in at least one of reduction of tumor size, reduction of tumor growth, inhibition of cancer metastases, inhibition of tumor recurrence, and cancer cell death.
  • 13. The drug combination of claim 10, wherein the heteroarotinoid is selected from SHetA2, SHetA3, SHetA4, SHetC2, SHetD3, SHetD4, SHetD5, SHet50, SHet65, SHet100, OHet72, NHet17, NHet86, and NHet90.
  • 14. The drug combination of claim 10, wherein the second compound is an Azabicyclooctan-3-one derivative selected from PRIMA-1met and PRIMA-1.
  • 15. The drug combination of claim 10, wherein the second compound is a CDK4/6 inhibitor.
  • 16. The drug combination of claim 15, wherein the CDK4/6 inhibitor is selected from the group consisting of Palbociclib, Abemaciclib, Ribociclib, Narazaciclib (ON123300), Dalpiciclib, Dinaciclib, Milciclib, Seliciclib, Wang-4d, and Wang-4e..
  • 17. The drug combination of claim 10, wherein the heteroarotinoid compound and a second compound are selected from the group consisting of the drug combinations in Tables 10-13.
  • 18. The drug combination of claim 10, wherein the heteroarotinoid comprises a dosage in a range of 1 mg/kg to 100 mg/kg, and the second compound comprises a dosage in a range of 1 mg/kg to 100 mg/kg.
  • 19. The drug combination of claim 10, wherein the heteroarotinoid and the second compound comprise a weight-to-weight ratio in a range of 50:1 to 1:50.
  • 20. A kit or commercial package, comprising a hetertoarotinoid, and a second compound selected from the group consisting of Azabicyclooctan-3-one derivatives, and CDK4/6 inhibitors, wherein the hetertoarotinoid and the second compound when administered conjointly provide for a synergistic effect against cancer cells compared to an effect of the heteroarotinoid alone and an effect of the second compound alone.
CROSS-REFERENCE TO RELATED APPLICATIONS

The present application is a continuation-in-part application which claims benefit to U.S. Ser. No. 17/276,032, filed Mar. 12, 2021, which claims priority to International Application No. PCT/US2019/050775, filed Sept. 12, 2019, which claims priority to 35 U.S.C. § 119(e) to U.S. Serial No. 62/730,345, filed Sept. 12, 2018. The present application also claims benefit to U.S. Ser. No. 18/145,334, filed Dec. 22, 2022. The entireties of each of the referenced applications are hereby expressly incorporated by reference herein.

GOVERNMENT SUPPORT

This invention was made with government support under grants CA196200 and CA200126 awarded by the National Institutes of Health. The government has certain rights in the invention.

Provisional Applications (1)
Number Date Country
62730345 Sep 2018 US
Continuation in Parts (1)
Number Date Country
Parent 17276032 Mar 2021 US
Child 18101627 US